EP3623364A1 - Prodrug compounds and their uses - Google Patents

Prodrug compounds and their uses Download PDF

Info

Publication number
EP3623364A1
EP3623364A1 EP19207646.1A EP19207646A EP3623364A1 EP 3623364 A1 EP3623364 A1 EP 3623364A1 EP 19207646 A EP19207646 A EP 19207646A EP 3623364 A1 EP3623364 A1 EP 3623364A1
Authority
EP
European Patent Office
Prior art keywords
optionally substituted
group
compound
alkyl
optionally
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP19207646.1A
Other languages
German (de)
French (fr)
Inventor
Lin Zhi
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ligand Pharmaceuticals Inc
Original Assignee
Ligand Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ligand Pharmaceuticals Inc filed Critical Ligand Pharmaceuticals Inc
Publication of EP3623364A1 publication Critical patent/EP3623364A1/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/661Phosphorus acids or esters thereof not having P—C bonds, e.g. fosfosal, dichlorvos, malathion or mevinphos
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/661Phosphorus acids or esters thereof not having P—C bonds, e.g. fosfosal, dichlorvos, malathion or mevinphos
    • A61K31/6615Compounds having two or more esterified phosphorus acid groups, e.g. inositol triphosphate, phytic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/662Phosphorus acids or esters thereof having P—C bonds, e.g. foscarnet, trichlorfon
    • A61K31/663Compounds having two or more phosphorus acid groups or esters thereof, e.g. clodronic acid, pamidronic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • A61K31/7072Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid having two oxo groups directly attached to the pyrimidine ring, e.g. uridine, uridylic acid, thymidine, zidovudine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D317/00Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms
    • C07D317/08Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3
    • C07D317/10Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3 not condensed with other rings
    • C07D317/14Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3 not condensed with other rings with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D317/18Radicals substituted by singly bound oxygen or sulfur atoms
    • C07D317/20Free hydroxyl or mercaptan
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D317/00Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms
    • C07D317/08Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3
    • C07D317/10Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3 not condensed with other rings
    • C07D317/14Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3 not condensed with other rings with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D317/18Radicals substituted by singly bound oxygen or sulfur atoms
    • C07D317/24Radicals substituted by singly bound oxygen or sulfur atoms esterified
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/04Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/06Phosphorus compounds without P—C bonds
    • C07F9/08Esters of oxyacids of phosphorus
    • C07F9/09Esters of phosphoric acids
    • C07F9/091Esters of phosphoric acids with hydroxyalkyl compounds with further substituents on alkyl
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/06Phosphorus compounds without P—C bonds
    • C07F9/08Esters of oxyacids of phosphorus
    • C07F9/09Esters of phosphoric acids
    • C07F9/098Esters of polyphosphoric acids or anhydrides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/655Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having oxygen atoms, with or without sulfur, selenium, or tellurium atoms, as the only ring hetero atoms
    • C07F9/65515Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having oxygen atoms, with or without sulfur, selenium, or tellurium atoms, as the only ring hetero atoms the oxygen atom being part of a five-membered ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6564Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms
    • C07F9/6571Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms having phosphorus and oxygen atoms as the only ring hetero atoms
    • C07F9/657154Cyclic esteramides of oxyacids of phosphorus
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6564Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms
    • C07F9/6571Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms having phosphorus and oxygen atoms as the only ring hetero atoms
    • C07F9/657163Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms having phosphorus and oxygen atoms as the only ring hetero atoms the ring phosphorus atom being bound to at least one carbon atom
    • C07F9/657181Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms having phosphorus and oxygen atoms as the only ring hetero atoms the ring phosphorus atom being bound to at least one carbon atom the ring phosphorus atom and, at least, one ring oxygen atom being part of a (thio)phosphonic acid derivative
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6564Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms
    • C07F9/6571Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms having phosphorus and oxygen atoms as the only ring hetero atoms
    • C07F9/6574Esters of oxyacids of phosphorus
    • C07F9/65744Esters of oxyacids of phosphorus condensed with carbocyclic or heterocyclic rings or ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6564Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms
    • C07F9/6571Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms having phosphorus and oxygen atoms as the only ring hetero atoms
    • C07F9/6574Esters of oxyacids of phosphorus
    • C07F9/65746Esters of oxyacids of phosphorus the molecule containing more than one cyclic phosphorus atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6564Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms
    • C07F9/6581Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms having phosphorus and nitrogen atoms with or without oxygen or sulfur atoms, as ring hetero atoms
    • C07F9/6584Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms having phosphorus and nitrogen atoms with or without oxygen or sulfur atoms, as ring hetero atoms having one phosphorus atom as ring hetero atom
    • C07F9/65842Cyclic amide derivatives of acids of phosphorus, in which one nitrogen atom belongs to the ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6564Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms
    • C07F9/6581Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms having phosphorus and nitrogen atoms with or without oxygen or sulfur atoms, as ring hetero atoms
    • C07F9/6584Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms having phosphorus and nitrogen atoms with or without oxygen or sulfur atoms, as ring hetero atoms having one phosphorus atom as ring hetero atom
    • C07F9/65842Cyclic amide derivatives of acids of phosphorus, in which one nitrogen atom belongs to the ring
    • C07F9/65844Cyclic amide derivatives of acids of phosphorus, in which one nitrogen atom belongs to the ring the phosphorus atom being part of a five-membered ring which may be condensed with another ring system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6564Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms
    • C07F9/6581Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms having phosphorus and nitrogen atoms with or without oxygen or sulfur atoms, as ring hetero atoms
    • C07F9/6584Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms having phosphorus and nitrogen atoms with or without oxygen or sulfur atoms, as ring hetero atoms having one phosphorus atom as ring hetero atom
    • C07F9/65842Cyclic amide derivatives of acids of phosphorus, in which one nitrogen atom belongs to the ring
    • C07F9/65846Cyclic amide derivatives of acids of phosphorus, in which one nitrogen atom belongs to the ring the phosphorus atom being part of a six-membered ring which may be condensed with another ring system
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • compositions and methods in the field of medicine and chemistry are disclosed. Some of the disclosed embodiments are directed to medicinal prodrug compounds, medicinal compositions, as well as processes for their preparation and methods of their use. Some embodiments include prodrug compounds of acid/alcohol derivatives, their preparation and their uses. In some embodiments, such prodrug compounds are useful to selectively deliver the acid/alcohol derivatives to the liver.
  • Prodrugs are frequently used to improve certain properties of pharmacological agents for a preferred route of administration, including physicochemical, biopharmaceutical or pharmacokinetic properties.
  • Certain prodrugs also called soft drugs
  • tissue selective activation or deactivation to achieve therapeutic advantages (See J. Rautio, et al. Nature Reviews Drug Discovery 7: 255-270 (2008 )).
  • cyclic phosphate, phosphonate, phosphonamidate, and phosphoramidate prodrugs are disclosed in US Patent No. 6,312,662 and US Patent No. 7,205,404 and designed for liver-targeting of pharmacological agents. These prodrugs are activated by liver cytochrome P450 enzymes CYP3As that are predominantly expressed in the target tissue and designed to achieve the selective delivery of pharmacological agents to the liver. Since the prodrugs are not active outside the liver, the liver-targeting strategy reduces any pharmacological or toxicological effects of a biologically active agent outside the targeting tissue.
  • liver-targeting strategy significantly improves patient benefit/risk ratio of a pharmacological agent (e.g. see M. D. Erion, et al. J Pharm Exp Ther 312:554-60 (2005 )).
  • a pharmacological agent e.g. see M. D. Erion, et al. J Pharm Exp Ther 312:554-60 (2005 )
  • Example activation of these cyclic phosph(on)ate and phosphoramidate compounds are illustrated below:
  • the latter is rapidly conjugated with glutathione (GSH) that exists in millimole levels in the liver to yield the conjugate byproduct.
  • GSH glutathione
  • liver-targeted property e.g. see X. J. Zhou, et al. 2009 EASL meeting poster #966 .
  • the liver-targeting effects of these agents are based on liver first-pass metabolism of an orally administered agent and the liver-targeting efficiency varies widely, depending upon the pharmacokinetic property of the agent, and are not as efficient as the Cyp3A activated prodrugs.
  • Novel prodrug compounds of acid/alcohol derivatives such as phosphates, phosphonates, phosphonamidates, phosphoramidates, carboxylates, phenolates, and alkoxylates, their preparation and their uses are described.
  • Some embodiments are related to novel prodrug compounds that do not generate a vinyl keto reactive intermediate in the activation process.
  • Some embodiments are directed to the use of the prodrugs to enhance oral drug delivery.
  • Another aspect includes the use of prodrugs to treat diseases that benefit from enhanced drug distribution to the liver and like tissues and cells, including but not limited to hepatitis, cancer, liver fibrosis, fatty liver, malaria, other viral and parasitic infections, and metabolic, cardiovascular, and/or hormonal diseases where the liver is involved in the production and/or the homeostasis control of the biochemical end products, e.g. glucose, cholesterol, fatty acids, bile acids, triglycerides, lipoproteins, apolipoproteins, and sex hormone-binding globulin (SHBG).
  • diseases include diabetes, hyperlipidemia, atherosclerosis, obesity and the like.
  • prodrugs are used to prolong pharmacodynamic half-life of a drug.
  • the prodrug methodology can be used to achieve sustained delivery of the parent drug.
  • prodrugs are used to increase the therapeutic index of the drug.
  • the prodrugs are useful in the delivery of diagnostic imaging agents to the liver.
  • Some embodiments relate to a compound of Formula I: wherein:
  • the compound is a compound of Formula Ia: wherein:
  • the compound is a compound of Formula Ib: wherein:
  • the compound is a compound of Formula Ic: wherein:
  • the compound is a compound of Formula Id or Ie: wherein:
  • Some embodiments relate to a compound of Formula II: wherein:
  • Some embodiments relate to a compound of Formula III: wherein:
  • Some embodiments relate to a compound of Formula IV: wherein:
  • Some embodiments relate to a compound of Formula V: wherein:
  • Some embodiments relate to a compound of Formula VI: wherein:
  • Some embodiments relate to a compound of Formula VII: wherein:
  • the compound is selected from the group consisting of: and a stereoisomer or a pharmaceutically acceptable salt thereof.
  • M is a nucleoside antiviral or anticancer agent.
  • M is a lipid modulator.
  • M is selected from the group consisting of HMG-CoA reductase inhibitor, a selective thyroid hormone receptor modulator, a peroxisome proliferator-activated receptor modulator, a fibrate, a nicotinic acid, a bile acid, and a fatty acid.
  • M is a glucose modulator
  • M is selected from the group consisting of a peroxisome proliferator-activated receptor modulator, a glucose biosynthesis inhibitor, and a dipeptidyl peptidase 4 inhibitor.
  • M is a nuclear hormone receptor modulator.
  • Some embodiments relate to a pharmaceutical composition
  • a pharmaceutical composition comprising any of the above compounds and a pharmaceutically acceptable excipient.
  • Some embodiments relate to a method of treating a disease, disorder or condition comprising administering an effective amount of any of the above compounds to a subject in need thereof.
  • the disease, disorder or condition is a disease, disorder or condition of the liver.
  • the disease, disorder or condition is a metabolic, cardiovascular or hormonal disease in which the liver is involved in the production and/or the homeostasis control of the biochemical end products of the disease, disorder or condition.
  • the disease, disorder or condition is selected from the group consisting of hepatitis, liver cancer, liver fibrosis, fatty liver, malaria, viral infection, parasitic infection, diabetes, hyperlipidemia, atherosclerosis, obesity, dyslipidemia, hyperglycemia and a hormonal condition.
  • Some embodiments relate to a method of treating a liver disease comprising administering an effective amount of any of the above compounds to a subject in need thereof, wherein M is a nucleoside antiviral or anticancer agent.
  • Some embodiments relate to a method of treating dyslipidemia comprising administering to a subject in need thereof an effective amount of any of the above compounds, wherein M is a lipid modulator.
  • M is selected from the group consisting of HMG-CoA reductase inhibitor, a selective thyroid hormone receptor modulator, peroxisome proliferator-activated receptor modulator, a fibrate, nicotinic acid, a bile acid, and a fatty acid.
  • Some embodiments relate to method of treating hyperglycemia comprising administering to a subject in need thereof, an effective amount of any of the above compounds, wherein M is a glucose modulator.
  • M is selected from the group consisting of peroxisome proliferator-activated receptor modulator, glucose biosynthesis inhibitor, and dipeptidyl peptidase 4 inhibitor.
  • Some embodiments relate to a method of treating a hormonal condition comprising administering to a subject in need thereof, an effective amount of any of the above compounds, wherein M is a nuclear hormone receptor modulator.
  • Some embodiments further comprise administering an effective amount of at least one additional therapeutic agent to the subject in need thereof.
  • Some embodiments relate to a method of delivering a diagnostic imaging agent to the liver of a subject in need thereof, comprising administering to the subject an effective amount of any of the above compounds.
  • the subject is a mammal.
  • the subject is human.
  • Some embodiments relate to a method of inhibiting viral replication in a cell comprising contacting the cell with any of the above compounds.
  • Some embodiments relate to a method of intervening in a molecular pathway or modulating a target in a cell comprising contacting the cell with any of the above compounds.
  • the cell is in vivo.
  • the cell is ex vivo.
  • the cell is a hepatocyte.
  • the cell is mammalian.
  • the cell is human.
  • the present embodiments are directed to compositions and methods related to novel prodrug compounds of biologically active acid/alcohol derivatives such as phosphates, phosphonates, phosphonamidates, phosphoramidates, carboxylates, phenolates, and alkoxylates, their preparation and their uses.
  • novel prodrug compounds of biologically active acid/alcohol derivatives such as phosphates, phosphonates, phosphonamidates, phosphoramidates, carboxylates, phenolates, and alkoxylates, their preparation and their uses.
  • Some embodiments relate to a compound of Formula I: wherein:
  • the compound is a compound of Formula Ia: wherein:
  • the compound is a compound of Formula Ib: wherein:
  • the compound is a compound of Formula Ic: wherein:
  • the compound is a compound of Formula Id or Ie: wherein:
  • Some embodiments relate to a compound of Formula IIa or IIb: wherein:
  • Some embodiments relate to a compound of Formula III: wherein:
  • Some embodiments relate to a compound of Formula IV: wherein:
  • Some embodiments relate to a compound of Formula Va, Vb, or Vc: wherein:
  • Some embodiments relate to a compound of Formula VIa, VIb or VIc: wherein:
  • Some embodiments relate to a compound of Formula VII: wherein:
  • CYP3A4 is expressed in the liver in a level much higher than other tissues ( DeWaziers et al. J Pharm Exp Ther 253:387 (1990 )).
  • Prodrug compounds of Formula I-VII are predominantly activated via CYP3A4 in the liver.
  • the prodrug compounds of Formulae I-VII have high efficiency in liver-targeting via selective delivery of biologically active agents to the liver.
  • the prodrugs are used to increase the therapeutic index of the drug, since the prodrug compounds of Formulae I-VII may not be active or may be less active outside the liver.
  • Certain drugs of phosph(on)ate derivatives are highly charged compounds that have generally poor oral bioavailability due to poor absorption in the gastrointestinal tract. Certain drugs are highly lipophilic compounds that have generally poor oral bioavailability due to poor absorption in the gastrointestinal tract. In some embodiments, the prodrug compounds of Formulae I-VII have oral bioavailability superior to the parent drugs/agents.
  • the compounds are used to treat diseases that benefit from enhanced drug distribution to the liver and like tissues and cells, including but not limited to diseases in the liver, such as hepatitis, liver cancer, liver fibrosis, fatty liver, malaria, other viral and parasitic infections, and metabolic, cardiovascular, and/or hormonal diseases where the liver is involved in the production and/or the homeostasis control of the biochemical end products, e.g.
  • glucose diabetes
  • cholesterol fatty acids
  • bile acids fatty acids
  • triglycerides hyperlipidemia
  • lipoproteins lipoproteins
  • apolipoproteins sex hormone-binding globulin (SHBG).
  • SHBG sex hormone-binding globulin
  • the disclosed prodrugs are used to prolong pharmacodynamic half-life of the drug.
  • the disclosed prodrug methodology can be used to achieve sustained delivery of the parent drug.
  • a method of making these prodrugs is described.
  • the prodrugs are also useful in the delivery of diagnostic imaging agents to the liver or other tissues.
  • Some compounds of Formulae I-VII have asymmetric centers where the stereochemistry is unspecified, and the diastereomeric mixtures of these compounds are included, as well as the individual stereoisomers when referring to a compound of Formulae I-VII generally.
  • compositions and methods provided herein include a pharmaceutical composition comprising a compound provided herein and a pharmaceutically acceptable carrier.
  • Some embodiments also include administering an effective amount of a second or multiple therapeutic agents in combination with a compound provided herein to the subject in need thereof.
  • Some embodiments of the compounds, compositions and methods provided herein include a method of treating a liver disease comprising administering an effective amount of a compound provided herein where M is a nucleoside antiviral or anticancer agent to a subject in need thereof.
  • Some embodiments of the compounds, compositions and methods provided herein include a method of treating dyslipidemia comprising administering an effective amount of a compound provided herein where M is a lipid modulator such as a HMG-CoA reductase inhibitor, a selective thyroid hormone receptor modulator, peroxisome proliferator-activated receptor modulator, a fibrate, nicotinic acid, and an omega-3 fatty acid to a subject in need thereof.
  • M is a lipid modulator such as a HMG-CoA reductase inhibitor, a selective thyroid hormone receptor modulator, peroxisome proliferator-activated receptor modulator, a fibrate, nicotinic acid, and an omega-3 fatty acid
  • Some embodiments of the compounds, compositions and methods provided herein include a method of treating hyperglycemia comprising administering an effective amount of a compound provided herein where M is a glucose modulator such as peroxisome proliferator-activated receptor modulator, glucose biosynthesis inhibitor, and dipeptidyl peptidase 4 inhibitor to a subject in need thereof.
  • M is a glucose modulator such as peroxisome proliferator-activated receptor modulator, glucose biosynthesis inhibitor, and dipeptidyl peptidase 4 inhibitor
  • Some embodiments of the compounds, compositions and methods provided herein include a method of treating a hormonal condition comprising administering an effective amount of a compound provided herein where M is a nuclear hormone receptor modulator to a subject in need thereof.
  • the subject is mammalian.
  • the subject is human.
  • Some embodiments of the compounds, compositions and methods provided herein include a method of testing a compound in a cell comprising contacting the cell with the compound of the claims.
  • the cell is in vivo.
  • the cell is ex vivo.
  • the cell is a hepatocyte.
  • the cell is mammalian.
  • the cell is human.
  • Some embodiments of the compounds, compositions and methods provided herein include use of a compound provided herein in the treatment of a disease in the liver.
  • Some embodiments include the use of a compound provided herein in combination with additional therapeutic agent(s) for the treatment of a disease in the liver.
  • Some embodiments of the compounds, compositions and methods provided herein include use of a compound provided herein in the treatment of a disease or condition by intervening in a molecular pathway in the liver.
  • Some embodiments include the use of a compound provided herein in combination with additional therapeutic agent(s) for the treatment of a disease or condition by intervening in a molecular pathway in the liver.
  • ranges and amounts can be expressed as “about” a particular value or range. “About” also includes the exact amount. Hence “about 10%” means “about 10%” and also “10%.”
  • an optionally substituted group means that the group is unsubstituted or is substituted.
  • compositions comprising "a therapeutic agent” includes compositions with one or a plurality of therapeutic agents.
  • alkyl refers to saturated aliphatic groups including straight-chain, branched chain and cyclic groups, up to and including 10 carbon atoms. Suitable alkyl groups include methyl, ethyl, n-propyl, isopropyl, and cyclopropyl. The alkyl group may be optionally substituted with 1-3 substituents.
  • optionally substituted or “substituted” includes groups substituted by one to four substituents, independently selected from lower alkyl, lower aryl, lower aralkyl, lower cyclic alkyl, lower heterocycloalkyl, hydroxy, lower alkoxy, lower aryloxy, perhaloalkoxy, aralkoxy, lower heteroaryl, lower heteroaryloxy, lower heteroarylalkyl, lower heteroaralkoxy, azido, amino, halogen, lower alkylthio, oxo, lower acylalkyl, lower carboxy esters, carboxyl, carboxamido, nitro, lower acyloxy, lower aminoalkyl, lower alkylaminoaryl, lower alkylaryl, lower alkylaminoalkyl, lower alkoxyaryl, lower arylamino, lower aralkylamino, lower alkylsulfonyl, lower carboxamidoalkylaryl, lower carboxamidoary
  • Substituted aryl and “substituted heteroaryl” refers to aryl and heteroaryl groups substituted with 1-6 substituents. These substituents are selected from the group consisting of lower alkyl, lower alkoxy, lower perhaloalkyl, halogen, hydroxy, cyano, and amino.
  • heteroalkyl refer to alkyl groups containing at least one heteroatom, such as 1 to 3 heteroatoms. Suitable heteroatoms include oxygen, sulfur, and nitrogen.
  • heteroacyl refer to -C(O)-heteroalkyl groups.
  • acyloxy refers to -OC(O)R where R is alkyl, or heteroalkyl.
  • alkoxy or “alkyloxy” refers to OR where R is alkyl, or heteroalkyl, all optionally substituted.
  • amino refers to NRR' where R and R' are each independently selected from hydrogen, alkyl, aryl, aralkyl and heterocycloalkyl, all except H are optionally substituted; and R and R' can form a cyclic ring system.
  • 'acylamino refers to -NRC(O)R' where R and R' are each independently selected from H, alkyl, or heteroalkyl.
  • halogen or halo refers to F, Cl, Br and I.
  • haloalkyl refer to alkyl groups containing at least one halogen, in a further aspect are 1 to 3 haloatoms. Suitable haloatoms include F, Cl, and Br.
  • haloheteroalkyl refer to alkyl groups containing at least one halogen and one heteroatom.
  • haloacyl refer to -C(O)-haloalkyl groups.
  • haloheteroacyl refer to -C(O)-haloheteroalkyl groups.
  • alkenyl refers to unsaturated groups which have 2 to 12 atoms and contain at least one carbon carbon double bond and includes straight chain, branched chain and cyclic groups. Alkenyl groups may be optionally substituted. Suitable alkenyl groups include allyl.
  • alkynyl refers to unsaturated groups which have 2 to 12 atoms and contain at least one carbon carbon triple bond and includes straight chain, branched chain and cyclic groups. Alkynyl groups may be optionally substituted. Suitable alkynyl groups include ethynyl.
  • aminoalkyl refers to the group NR 2 -alkyl where R is selected from H, alkyl, aryl, aralkyl, and heterocycloalkyl.
  • alkylthio refers to the group alkyl-S-.
  • perhalo refers to groups wherein every C-H bond has been replaced with a C-halo bond on an aliphatic or aryl group.
  • Suitable perhaloalkyl groups include CF 3 and CFCl 2 .
  • aryl refers to an aromatic group wherein each of the atoms forming the ring is a carbon atom.
  • Aryl rings may be formed by five, six, seven, eight, nine, or more than nine carbon atoms.
  • Aryl groups may be optionally substituted. Examples of aryl groups include, but are not limited to phenyl, naphthalenyl, phenanthrenyl, anthracenyl, tetralinyl, fluorenyl, indenyl, and indanyl.
  • heteroaryl refers to an aromatic group wherein at least one atom forming the aromatic ring is a heteroatom. Heteroaryl rings may be formed by three, four, five, six, seven, eight, nine, or more than nine atoms. Heteroaryl groups may be optionally substituted. Examples of heteroaryl groups include, but are not limited to, aromatic C 3-8 heterocyclic groups comprising one oxygen or sulfur atom or up to four nitrogen atoms, or a combination of one oxygen or sulfur atom and up to two nitrogen atoms, and their substituted as well as benzo- and pyrido-fused derivatives, for example, connected via one of the ring-forming carbon atoms.
  • heteroaryl groups are optionally substituted with one or more substituents, independently selected from halo, hydroxy, amino, cyano, nitro, alkylamido, acyl, C 1-6 -alkoxy, C 1-6 -alkyl, C 1-6 -hydroxyalkyl, C 1-6 -aminoalkyl, C 1-6 -alkylamino, alkylsulfenyl, alkylsulfinyl, alkylsulfonyl, sulfamoyl, or trifluoromethyl.
  • substituents independently selected from halo, hydroxy, amino, cyano, nitro, alkylamido, acyl, C 1-6 -alkoxy, C 1-6 -alkyl, C 1-6 -hydroxyalkyl, C 1-6 -aminoalkyl, C 1-6 -alkylamino, alkylsulfenyl, alkylsulfinyl, alkylsulf
  • heteroaryl groups include, but are not limited to, unsubstituted and mono- or di-substituted derivatives of furan, benzofuran, thiophene, benzothiophene, pyrrole, pyridine, indole, oxazole, benzoxazole, isoxazole, benzisoxazole, thiazole, benzothiazole, isothiazole, imidazole, benzimidazole, pyrazole, indazole, tetrazole, quinoline, isoquinoline, pyridazine, pyrimidine, purine and pyrazine, furazan, 1,2,3-oxadiazole, 1,2,3-thiadiazole, 1,2,4-thiadiazole, triazole, benzotriazole, pteridine, phenoxazole, oxadiazole, benzopyrazole, quinolizine, cinnoline, phthalazin
  • the substituents are halo, hydroxy, cyano, O-C 1-6 -alkyl, C 1-6 -alkyl, hydroxy-C 1-6 -alkyl, and amino-C 1-6 -alkyl.
  • aryloxy refers to -O-aryl.
  • heteroaryloxy refers to -O-heteroaryl
  • terapéuticaally effective amount means an amount of a compound or a combination of compounds that partially or fully ameliorates, attenuates or eliminates one or more of the symptoms of a particular disease or condition or prevents, modifies, or delays the onset of one or more of the symptoms of a particular disease or condition. Such amount can be administered as a single dosage or can be administered according to a regimen, whereby it is effective. Repeated administration may be needed to achieve a desired result (e.g., treatment of the disease and/or condition).
  • salts of compounds of Formulae I-VII and its prodrugs derived from the combination of a compound of the present embodiments and an organic or inorganic acid or base include acetic acid, adipic acid, benzenesulfonic acid, (+)-7,7-dimethyl-2-oxobicyclo[2.2.1]heptane-1-methanesulfonic acid, citric acid, 1,2-ethanedisulfonic acid, dodecyl sulfonic acid, fumaric acid, glucoheptonic acid, gluconic acid, glucuronic acid, hippuric acid, hydrochloride hemiethanolic acid, HBr, HCl, HI, 2-hydroxyethanesulfonic acid, lactic acid, lactobionic acid, maleic acid, methanesulfonic acid, methylbromide acid, methyl sulfuric acid, 2-naphthalenesulfonic acid, nitric
  • Suitable bases include NaOH, KOH, Ca(OH) 2 , Zn(OH) 2 , Mg(OH) 2 , diethylamine, ethanolamine, diethanolamine, choline, lysine, meglumine, benzathine, and tromethamine.
  • spacer refers to an atom or group of atoms that separate two or more groups from one another by a desired number of atoms. For example, in some embodiments, it may be desirable to separate two or more groups by one, two, three, four, five, six, or more than six atoms. In such embodiments, any atom or group of atoms may be used to separate those groups by the desired number of atoms. Spacers are optionally substituted. In some embodiments, a spacer comprises saturated or unsaturated alkyls, heteroalkyls and/or haloalkyls. In some embodiments, a spacer comprises atoms that are part of a ring.
  • haloalkyl can include one or more of the same or different halogens.
  • haloalkyl includes each of the substituents CF 3 , CHF 2 and CH 2 F.
  • spacers are provided.
  • 2 atom spacers include, but are not limited to, the following: where A and B represent groups which are separated by the desired number of atoms.
  • Examples of 3 atom spacers include, but are not limited to, the following: where A and B represent groups which are separated by the desired number of atoms.
  • patient refers to an animal being treated including a mammal, such as a dog, a cat, a cow, a horse, a sheep, and a human.
  • a mammal such as a dog, a cat, a cow, a horse, a sheep, and a human.
  • the patient is a mammal, either male or female.
  • the patient is a male or female human.
  • prodrug refers to any compound that when administered to a biological system generates a biologically active compound as a result of spontaneous chemical reaction(s), enzyme catalyzed chemical reaction(s), and/or metabolic chemical reaction(s), or a combination of each.
  • Standard prodrugs are formed using groups attached to functionality, e.g. HO-, HS-, HOOC-, HOOPR 2 -, associated with the drug, that cleave in vivo.
  • Standard prodrugs include but are not limited to carboxylate esters where the group is alkyl, aryl, aralkyl, acyloxyalkyl, alkoxycarbonyloxyalkyl as well as esters of hydroxyl, thiol and amines where the group attached is an acyl group, an alkoxycarbonyl, aminocarbonyl, phosphate or sulfate.
  • the groups illustrated are examples, not exhaustive, and one skilled in the art could prepare other known varieties of prodrugs.
  • Such prodrugs of the compounds of Formula I fall within this scope.
  • Prodrugs must undergo some form of a chemical transformation to produce the compound that is biologically active or is a precursor of the biologically active compound.
  • the prodrug is biologically active, usually less than the drug itself, and serves to improve drug efficacy or safety through improved oral bioavailability, pharmacodynamic half-life, etc.
  • Prodrug forms of compounds may be utilized, for example, to improve bioavailability, improve subject acceptability such as by masking or reducing unpleasant characteristics such as bitter taste or gastrointestinal irritability, alter solubility such as for intravenous use, provide for prolonged or sustained release or delivery, improve ease of formulation, or provide site specific delivery of the compound.
  • stereoisomer refers to the relative or absolute spatial relationship of the R group(s) and the substituent attached to the phosphorus atom via an exocyclic single bond on the 2-oxo-phosphorus prodrug ring, and refers to individual or any combination of the individual isomers such as a racemic mixture and a diastereomeric mixture.
  • R group is attached to the carbon atom in the ring
  • stereoisomers there are four stereoisomers.
  • the structures A, B, C, and D below show four possible individual isomers. Structures A and D (or B and C) are a pair of two enantiomers (or called optical isomers).
  • two R groups attached to two different carbon atoms in the ring there are eight possible stereoisomers.
  • liver refers to the liver organ.
  • liver specificity refers to the ratio: drug or a drug metabolite in liver tissue drug or a drug metabolite in blood or another tissue
  • the ratio can be determined by measuring tissue levels at a specific time or may represent an AUC (area under a curve) based on values measured at three or more time points.
  • liver specificity refers to an increase in liver specificity ratio in animals treated with the prodrug relative to animals treated with the parent drug.
  • Compounds disclosed in US Patent No. 8,063,025 , US Patent No. 7,666,855 , and PCT Pub. No. WO2009/073506 are designed for the liver-specific delivery of nucleosides for the treatment of HCV patients and take advantage of a cytochrome P450 enzyme that is mainly expressed in the liver.
  • the term "enhanced oral bioavailability" refers to an increase of at least about 50% of the absorption of the dose of the parent drug. In an additional aspect, the increase in oral bioavailability of the prodrug (compared to the parent drug) is at least about 100%, or a doubling of the absorption. Measurement of oral bioavailability usually refers to measurements of the prodrug, drug, or drug metabolite in blood, plasma, tissues, or urine following oral administration compared to measurements following parenteral administration.
  • therapeutic index refers to the ratio of the dose of a drug or prodrug that produces a therapeutically beneficial response relative to the dose that produces an undesired response such as death, an elevation of markers that are indicative of toxicity, and/or pharmacological side effects.
  • sustained delivery refers to an increase in the period in which there is a prolongation of therapeutically-effective drug levels due to the presence of the prodrug.
  • treating includes inhibiting the disease (slowing or arresting or partially arresting its development), providing relief from the symptoms or side effects of the disease (including palliative treatment), and/or relieving the disease (causing regression of the disease).
  • biological agent refers to a compound that has biological activity or that has molecular properties that can be used for diagnosis purpose, such as a compound carrying a radioactive isotope or a heavy atom.
  • molecular pathway refers to a series of molecular events in tissues such as a receptor modulating sequence or a biosynthesis sequence that is involved in physiological or pathophysiological functions of a living animal.
  • vinyl ketone reactive intermediate refers to compounds of the structure below that are chemically reactive to generate a covalent bond with a molecular entity in the tissues or cell, where R is H, alkyl, aryl, or heteroaryl.
  • the disclosed compounds may be used alone or in combination with other treatments. These compounds, when used in combination with other agents, may be administered as a daily dose or an appropriate fraction of the daily dose (e.g., bid). The compounds may be administered after a course of treatment by another agent, during a course of therapy with another agent, administered as part of a therapeutic regimen, or may be administered prior to therapy by another agent in a treatment program.
  • Examples of pharmaceutically acceptable salts include acetate, adipate, besylate, bromide, camsylate, chloride, citrate, edisylate, estolate, fumarate, gluceptate, gluconate, glucuronate, hippurate, hyclate, hydrobromide, hydrochloride, iodide, isethionate, lactate, lactobionate, maleate, mesylate, methylbromide, methylsulfate, napsylate, nitrate, oleate, palmoate, phosphate, polygalacturonate, stearate, succinate, sulfate, sulfosalicylate, tannate, tartrate, terphthalate, tosylate, triethiodide, sodium, potassium, calcium, zinc, magnesium, diethylamine, ethanolamine, diethanolamine, cholinate, lysine, meglumine, benzathine, and
  • compositions containing the active ingredient may be in any form suitable for the intended method of administration.
  • the compounds of a method and/or composition described herein can be provided via oral administration, rectal administration, transmucosal administration, intestinal administration, enteral administration, topical administration, transdermal administration, intrathecal administration, intraventricular administration, intraperitoneal administration, intranasal administration, intraocular administration and/or parenteral administration.
  • compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation. Tablets containing the active ingredient in admixture with non-toxic pharmaceutically acceptable excipient which are suitable for manufacture of tablets are acceptable.
  • excipients may be, for example, inert diluents, such as calcium or sodium carbonate, lactose, calcium or sodium phosphate; granulating and disintegrating agents, such as maize starch, or alginic acid; binding agents, such as starch, gelatin or acacia; and lubricating agents, such as magnesium stearate, stearic acid or talc. Tablets may be uncoated or may be coated by known techniques including microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax may be employed.
  • inert diluents such as calcium or sodium carbonate, lactose, calcium or sodium phosphate
  • granulating and disintegrating agents such as maize starch, or alginic acid
  • binding agents such as starch, ge
  • Formulations for oral use may be also presented as hard gelatin capsules where the active ingredient can be mixed with an inert solid diluent, for example calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient can be mixed with water or an oil medium, such as peanut oil, liquid paraffin or olive oil.
  • an inert solid diluent for example calcium phosphate or kaolin
  • an oil medium such as peanut oil, liquid paraffin or olive oil.
  • Formulations suitable for parenteral administration include aqueous and non-aqueous isotonic sterile injection solutions which may contain, for example, antioxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the formulations may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use.
  • Injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
  • unit dosage formulations contain a daily dose or unit, daily sub-dose, or an appropriate fraction thereof, of a drug. It will be understood, however, that the specific dose level for any particular patient will depend on a variety of factors including the activity of the specific compound employed; the age, body weight, general health, sex and diet of the individual being treated; the time and route of administration; the rate of excretion; other drugs which have previously been administered; and the severity of the particular disease undergoing therapy, as is well understood by those skilled in the art.
  • the following procedures for the preparation of the cyclic prodrug compounds illustrate the general procedures used to prepare the compounds which apply to phosphate, phosphonate, phosphoramidate, carboxylic acid, and alcohol containing drugs.
  • Prodrugs can be introduced at different stages of synthesis of a drug. In some embodiments, they are made at a later stage, because of the general sensitivity of these groups to various reaction conditions.
  • Optically pure prodrugs containing a single isomer at the phosphorus center can be made, for example, by separation of the diastereomers by a combination of column chromatography and/or crytallyzation, or by enantioselective synthesis of chiral activated phosph(on)ate intermediates.
  • Scheme I describes general strategies of synthesis of the cyclic phosph(on)ate prodrug compounds of Formula I.
  • the first strategy starts with treatment of a dihydroxyl compound of structure 1 where Z is not a heteroatom with phosphorus oxychloride to generate a cyclic phosphate of structure 2.
  • a coupling reaction of a cyclic phosphate chloride of structure 2 with an alcohol derivative compound of structure 3 provides the final compound of structure 4.
  • Z is a heteroatom
  • an alternative strategy can be used where the alcohol compound of structure 3 is converted to its monophosphate of structure 5 via a two-step sequence of phosphorylation with phosphorus oxychloride and acid hydrolysis by a resin.
  • the monophosphate of structure 5 is then treated with a dichloro compound of structure 6 in the presence of silver nitrate to give the final product of structure 4.
  • Scheme II describes general strategies of synthesis of cyclic phosphoramidate or phosphonamidate prodrug compounds of Formula I.
  • Treatment of the hydroxylamino compounds of structure 7 (where Z is not a heteroatom) with phosphorus oxychloride provides the cyclic phosphoramidate of structure 8.
  • a biological agent having a hydroxyl group of structure 3 is coupled with a prodrug intermediate of structure 8 to yield the final product of structure 9.
  • a chloroamino compound of structure 10 (where Z is a heteroatom) is treated with phosphorus oxychloride and then an ion-exchange resin sequentially to give the intermediate of structure 11 that undergoes an internal cyclization followed by chlorination to the cyclic phosphoramidate of structure 8.
  • Scheme III describes general synthetic procedures of the 2-oxodioxazaphinine prodrugs of Formula I.
  • a biological agent derivative of structure 12 is treated with phosphorus oxychloride to provide an intermediate of structure 13 that is then treated with a base in the presence of an aldehyde of structure 14 to give final product of structure 15.
  • the regioisomers of structure 15 are made from a different route where a compound of structure 16 is treated sequentially with diphenyl phosphate under basic a condition to give an intermediate of structure 17.
  • Base mediated internal cyclization of the intermediate of structure 17 affords the final product of structure 18.
  • Scheme IV describes general synthetic procedures of the prodrugs of Formula IIa.
  • Benzaldehyde derivative of structure 19 is treated with phosphorus oxychloride to provide an intermediate of structure 20 that is then converted to structure 21 in the presence of a acid chloride derivative of a biologic agent with zinc chloride as the catalyst.
  • Deprotection followed by treatment of a base affords the final product of structure 22.
  • Scheme V describes a procedure for preparing cyclic bisphosphonate prodrug compounds of Formula III via either an acid catalyzed condensation or a base catalyzed alkylation routes.
  • Condensation reaction of a bisphosphonate of structure 23 with an aldehyde of structure 24 in the presence of an acid catalyst affords a product of structure 25.
  • alkylation of a bisphosphonate of structure 23 with a halide compound of structure 26 in the presence of silver nitrate and/or a base provides the same product of structure 25.
  • Scheme VI describes general strategies of synthesis of the cyclic acetal prodrug compounds of Formula IV.
  • the dihydroxyl compound of structure 1 is condensed with an aldehyde of structure 2 in the presence of catalytic amount of acid to give a product of structure 3.
  • Aldehyde compound of structure 2 is prepared from the corresponding carboxylic acid by the standard procedure in the literature.
  • Some biologically active compounds of Formulae I-VII are prepared as outlined below. Some biologically nonactive compounds of Formulae I-VII are also prepared to demonstrate the synthetic methodologies as outlined below.
  • Compound 119 was prepared as a mixture of stereoisomers from Compound 118 and dimethyl chlorophosphate.
  • Compounds 120 and 121 were prepared as a mixture of stereoisomers from oleic acid, nicotinaldehyde, and glycerol. Briefly, nicotinaldehyde was condensed with glycerol in the presence of an acid catalyst to afford a mixture of 5-member-ring and 6-member-ring acetals that were treated with oleic chloride to form the corresponding ester Compounds 120 and 121. [M+H] + calcd for C 27 H 43 NO 4 : 446.32; found: 446.4.
  • Compound 122 was prepared from oleic acid, 2,2-dimethylpropane-1,3-diol, and phosphorus oxychloride. Briefly, 2,2-dimethylpropane-1,3-diol was treated with phosphorus oxychloride to form the corresponding cyclic chlorophosphate that was then reacted with oleic amide to provide Compound 122. [M+H] + calcd for C 23 H 44 NO 4 P: 430.30; found: 430.6.
  • Compound 123 was prepared as a mixture of stereoisomers from oleic acid, 2,2-dimethyl-1-phenylpropane-1,3-diol, and phosphorus oxychloride in a reaction sequence same as that of Compound 122. [M+H] + calcd for C 29 H 48 NO 4 P: 506.33; found: 506.4.
  • Compound 124 was prepared as a mixture of stereoisomers from oleic acid, 2,2-dimethyl-1-phenylpropane-1,3-diol, and phosphorus oxychloride. Briefly, oleic acid was converted to the acid chloride with treatment of oxalyl chloride in dichloromethane and then the oleic chloride was treated with paraldehyde and zinc chloride in acetonitrile at 60-65 °C to give 1-chloroethyl oleate.
  • Example A In Vitro Activation of Prodrug Analogues by Rat Liver Microsomes. Quantification by LC-MS/MS
  • Prodrug compounds are tested for activation to the active drug in reactions catalyzed by the microsomal fraction of rat liver.
  • Prodrug compounds are tested for activation by liver microsomes isolated from rats induced with dexamethasone to enhance CYP3A4 activity. Reactions are conducted in 0.1 M KH 2 PO 4 , pH 7.4, in the presence of 2 mM NADPH and liver microsomes (1 mg/mL). Reaction mixtures are incubated for 5 min. in an Eppendorf Thermomixer 5436 (37 °C, speed 6). Reactions are terminated by the addition of 1.5 volumes of methanol. The resulting extracts are clarified by centrifugation at 14,000 rpm in an Eppendorf microfuge (20 min.). The supernatants (200 ⁇ L) are evaporated under vacuum and heat to dryness.
  • the dried residue is reconstituted with 200 ⁇ L of water and the mixture is centrifuged for 10 min at 14,000 rpm. A mixture of 35 ⁇ L aliquot of supernatant and 35 ⁇ L of mobile phase A is analyzed by LC-MS/MS. The active compound is detected by using MS/MS mode and quantified based on comparison to a standard active compound.
  • Example B Active compound Accumulation in Hepatocytes Following Incubation with Prodrug compounds
  • Prodrug compounds are evaluated for their ability to generate active compounds in freshly isolated rat hepatocytes.
  • Hepatocytes are prepared from fed Sprague-Dawley rats (250-300g) according to the procedure of Berry and Friend ( Berry, M.N. Friend, D.S., J. Cell Biol. 43:506-520 (1969 )) as modified by Groen ( Groen, A.K. et al., Eur. J. Biochem 122:87-93 (1982 )).
  • Hepatocytes (20 mg/mL wet weight, >85% trypan blue viability) are incubated at 37 °C in 2 mL of Krebs-bicarbonate buffer containing 20 mM glucose, and 1 mg/mL BSA for 2 h in the presence of 1-250 ⁇ M a prodrug compound (from 25 mM stock solutions in DMSO). Following the incubation, 1600 ⁇ L aliquot of the cell suspension is centrifuged and 300 ⁇ L of acetonitrile is added to the pellet, vortexed and sonicated until the pellet broke down. Then 200 ⁇ L of water is added to make a 60% acetonitrile solution.
  • liver specificity of prodrugs is compared relative to their parent active compound in liver and other organs that could be targets of toxicity.
  • Nucleoside analogues and their prodrugs are administered at 5-20 mg/kg to fasted rats by oral gavage.
  • Plasma and portal vein concentrations of the active and prodrug are determined by HPLC-UV, and the liver, skeletal muscle, cardiac, kidney, small intestine, and other organ concentrations are measured by LC-MS using the standard chromatography method.
  • the results demonstrate the liver targeting of the prodrug compounds and provide evidence for improved safety of the prodrugs over that of the actives. This can occur solely by the liver targeting provided by the prodrug.
  • the terms “generally parallel” and “substantially parallel” refer to a value, amount, or characteristic that departs from exactly parallel by less than or equal to 15°, 10°, 5°, 3°, 1°, 0.1°, or otherwise.
  • the terms “generally perpendicular” and “substantially perpendicular” refer to a value, amount, or characteristic that departs from exactly perpendicular by less than or equal to 15°, 10°, 5°, 3°, 1°, 0.1°, or otherwise.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Epidemiology (AREA)
  • Biochemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Diabetes (AREA)
  • Nutrition Science (AREA)
  • Physiology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Virology (AREA)
  • Obesity (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Hematology (AREA)
  • Endocrinology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Saccharide Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicinal Preparation (AREA)

Abstract

Provided herein are prodrug compounds of formula (IV), their preparation and their uses, such as treating liver diseases or non-liver diseases via intervening in molecular pathways in the liver.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims benefit of U.S. Provisional Application No. 61/939,615, filed on February 13, 2014 , U.S. Provisional Application No. 61/988,101, filed on May 2, 2014 , and U.S. Provisional Application No. 61/988,118, filed on May 2, 2014 which are incorporated herein by reference in their entirety.
  • FIELD
  • Compositions and methods in the field of medicine and chemistry are disclosed. Some of the disclosed embodiments are directed to medicinal prodrug compounds, medicinal compositions, as well as processes for their preparation and methods of their use. Some embodiments include prodrug compounds of acid/alcohol derivatives, their preparation and their uses. In some embodiments, such prodrug compounds are useful to selectively deliver the acid/alcohol derivatives to the liver.
  • BACKGROUND
  • The following description of the background is provided to aid in understanding the invention, but is not admitted to be, or to describe, prior art.
  • Prodrugs are frequently used to improve certain properties of pharmacological agents for a preferred route of administration, including physicochemical, biopharmaceutical or pharmacokinetic properties. Certain prodrugs (also called soft drugs) are designed by tissue selective activation or deactivation to achieve therapeutic advantages (See J. Rautio, et al. Nature Reviews Drug Discovery 7: 255-270 (2008)).
  • Certain cyclic phosphate, phosphonate, phosphonamidate, and phosphoramidate prodrugs are disclosed in US Patent No. 6,312,662 and US Patent No. 7,205,404 and designed for liver-targeting of pharmacological agents. These prodrugs are activated by liver cytochrome P450 enzymes CYP3As that are predominantly expressed in the target tissue and designed to achieve the selective delivery of pharmacological agents to the liver. Since the prodrugs are not active outside the liver, the liver-targeting strategy reduces any pharmacological or toxicological effects of a biologically active agent outside the targeting tissue. As a result, once used to treat liver diseases or to treat diseases via intervening in molecular pathways in the liver, the liver-targeting strategy significantly improves patient benefit/risk ratio of a pharmacological agent (e.g. see M. D. Erion, et al. J Pharm Exp Ther 312:554-60 (2005)). Example activation of these cyclic phosph(on)ate and phosphoramidate compounds are illustrated below:
    Figure imgb0001
  • In the above example, the cyclic prodrugs (X = O or N) are oxidized by Cyp3A in the liver and undergo a ring opening and β-elimination sequence to provide the active drugs and an aryl vinyl ketone (Intermediate). The latter is rapidly conjugated with glutathione (GSH) that exists in millimole levels in the liver to yield the conjugate byproduct.
  • Certain oral available pharmaceutical agents have been described to have certain liver-targeted property (e.g. see X. J. Zhou, et al. 2009 EASL meeting poster #966). The liver-targeting effects of these agents are based on liver first-pass metabolism of an orally administered agent and the liver-targeting efficiency varies widely, depending upon the pharmacokinetic property of the agent, and are not as efficient as the Cyp3A activated prodrugs.
  • SUMMARY
  • Novel prodrug compounds of acid/alcohol derivatives such as phosphates, phosphonates, phosphonamidates, phosphoramidates, carboxylates, phenolates, and alkoxylates, their preparation and their uses are described. Some embodiments are related to novel prodrug compounds that do not generate a vinyl keto reactive intermediate in the activation process. Some embodiments are directed to the use of the prodrugs to enhance oral drug delivery. Another aspect includes the use of prodrugs to treat diseases that benefit from enhanced drug distribution to the liver and like tissues and cells, including but not limited to hepatitis, cancer, liver fibrosis, fatty liver, malaria, other viral and parasitic infections, and metabolic, cardiovascular, and/or hormonal diseases where the liver is involved in the production and/or the homeostasis control of the biochemical end products, e.g. glucose, cholesterol, fatty acids, bile acids, triglycerides, lipoproteins, apolipoproteins, and sex hormone-binding globulin (SHBG). Examples of such diseases include diabetes, hyperlipidemia, atherosclerosis, obesity and the like. In another aspect, prodrugs are used to prolong pharmacodynamic half-life of a drug. In some embodiments, the prodrug methodology can be used to achieve sustained delivery of the parent drug. In another aspect, prodrugs are used to increase the therapeutic index of the drug. In some embodiments, the prodrugs are useful in the delivery of diagnostic imaging agents to the liver. Some additional embodiments relate to a method of making prodrugs.
  • Some embodiments relate to a compound of Formula I:
    Figure imgb0002
    wherein:
    • R1 and R2 are each independently selected from the group consisting of H, M, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 acyloxy, an optionally substituted aryl, and an optionally substituted heteroaryl;
    • R3 is selected from the group consisting of H and an optionally substituted C1-C6 alkyl; or R3 optionally forms an optionally substituted ring with R2; or R3 together with R2 form a methylene or its derivative; or R3 together with R2 form an oxo (=O) or its derivative; or R3 optionally forms a bond with Z or Y' when Z or Y' is N;
    • M is a biological agent, or part of a biological agent or a prodrug of a biological agent;
    • X is selected from the group consisting of Cl, OR4, NR4R5, an optionally substituted C1-C6 alkyl, and M;
    • Y and Y' are each independently O or NR4; or Y' is CH2 or null;
    • Z is selected from the group consisting of O, NR5, CR8R6, C=O, C=NR7, and null; or Z is a 2-5 atom spacer selected from an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 heteroalkyl, an optionally substituted aryl, and an optionally substituted heteroaryl;
    • R4 is selected from the group of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
    • R5 is selected from the group consisting of H, M, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, and an optionally substituted C1-C6 acyl;
    • R6 is selected from the group consisting of F, Cl, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl; or R6 is H provided that R1 or R2 or R8 is connected to a cyclic core carbon atom through an oxygen-carbon (O-C) bond;
    • R7 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
    • R8 is selected from the group consisting of F, Cl, M, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl; or R8 is H provided that R1 or R2 is connected to a cyclic core carbon atom through an oxygen-carbon (O-C) bond;
    • provided that at least one of R1, R2, R5, R8, and X is M;
    • or a stereoisomer or a pharmaceutically acceptable salt thereof.
  • In some embodiments, the compound is a compound of Formula Ia:
    Figure imgb0003
    wherein:
    • R1 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, an optionally substituted aryl, and an optionally substituted heteroaryl;
    • R2 and R3 are each independently selected from the group consisting of H and an optionally substituted C1-C6 alkyl; or R2 and R3 optionally form an optionally substituted ring; or R2 and R3 optionally form a methylene or its derivative; or R2 and R3 optionally form an oxo or its derivative;
    • M is a biological agent, part of a biological agent or a prodrug of a biological agent;
    • Y and Y' are each independently O or NR4; or Y' is CH2;
    • Z is selected from the group consisting of O, NR5, CR8R6, C=O, C=NR7, and null; or Z is a 2-5 atom spacer selected from an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 heteroalkyl, an optionally substituted aryl, and an optionally substituted heteroaryl;
    • R4 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
    • R5 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, and an optionally substituted C1-C6 acyl;
    • R6 is selected from the group consisting of F, Cl, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl; or R6 is H provided that R1 or R2 or R8 is connected with the cyclic core carbon atom through an oxygen-carbon (O-C) bond;
    • R7 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
    • R8 is selected from the group consisting of F, Cl, M, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl; or R8 is H provided that R1 or R2 is connected to a cyclic core carbon atom through an oxygen-carbon (O-C) bond;
    • or a stereoisomer or a pharmaceutically acceptable salt thereof.
  • In some embodiments, the compound is a compound of Formula Ib:
    Figure imgb0004
    wherein:
    • R2 and R3 are independently selected from the group consisting of H and an optionally substituted C1-C6 alkyl; or R2 and R3 optionally form an optionally substituted ring; or R2 and R3 optionally form a methylene or its derivative; or R2 and R3 optionally form an oxo (=O) or its derivative;
    • M is a biological agent, part of a biological agent or a prodrug of a biological agent;
    • X is selected from the group consisting of Cl, an optionally substituted C1-C6 alkyl, NR4R5, and OR4;
    • Y and Y' are each independently O or NR4; or Y' is CH2 or null;
    • Z is selected from the group consisting of O, NR5, C(R6)2, C=O, C=NR7, and null; or Z is a 2-5 atom spacer selected from an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 heteroalkyl, an optionally substituted aryl, and an optionally substituted heteroaryl;
    • R4 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
    • R5 is selected from the group consisting of H, an optionally substituted Ci-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, and an optionally substituted C1-C6 acyl;
    • R6 is selected from the group consisting of F, Cl, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl; or R6 is H provided that M or R2 is connected with the cyclic core carbon atom through an oxygen-carbon (O-C) bond;
    • R7 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
    • or a stereoisomer or a pharmaceutically acceptable salt thereof.
  • In some embodiments, the compound is a compound of Formula Ic:
    Figure imgb0005
    wherein:
    • R1 is selected from the group consisting of H, an optionally substituted Ci-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, an optionally substituted aryl, and an optionally substituted heteroaryl;
    • R3 is selected from the group consisting of H and an optionally substituted C1-C6 alkyl; or R3 together with M form a methnylene derivative; or R3 optionally forms a bond with Z or Y' when Z or Y' is N;
    • M is a biological agent, part of a biological agent or a prodrug of a biological agent;
    • X is selected from the group consisting of Cl, OR4, NR4R5, and an optionally substituted C1-C6 alkyl;
    • Y and Y' are each independently O or NR4;
    • Z is selected from the group consisting of O, NR5, C(R6)2, C=O, C=NR7, and null; or Z is a 2-5 atom spacer selected from an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 heteroalkyl, an optionally substituted aryl, and an optionally substituted heteroaryl;
    • R4 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
    • R5 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, and an optionally substituted C1-C6 acyl;
    • R6 is selected from the group consisting of F, Cl, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl; or R6 is H provided that R1 or M is connected with the cyclic core carbon atom through an oxygen-carbon (O-C) bond;
    • R7 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
    • or a stereoisomer or a pharmaceutically acceptable salt thereof.
  • In some embodiments, the compound is a compound of Formula Id or Ie:
    Figure imgb0006
    wherein:
    • R2 and R3 are independently selected from the group consisting of H and an optionally substituted C1-C6 alkyl; or R3 together with M or R2 form a methylene derivative; or R3 together with M or R2 form an optionally substituted ring; or R3 together with R2 form an oxo (=O) or its derivative; or R3 optionally forms a bond with Z or Y' when Z or Y' is N;
    • M is a biological agent, part of a biological agent or a prodrug of a biological agent;
    • X is selected from the group consisting of Cl, OR4, and an optionally substituted C2-C6 alkyl;
    • Y and Y' are each independently O or N; or Y' is CH2;
    • Z is selected from the group consisting of O and NR5;
    • R4 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
    • R5 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, and an optionally substituted C1-C6 acyl;
    • or a stereoisomer or a pharmaceutically acceptable salt thereof.
  • Some embodiments relate to a compound of Formula II:
    Figure imgb0007
    Figure imgb0008
    wherein:
    • R21 is selected from the group consisting of H, M, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 acyloxy, an optionally substituted aryl, and an optionally substituted heteroaryl;
    • M is a biological agent, or part of a biological agent or a prodrug of a biological agent;
    • Q is an optionally substituted aryl or an optionally substituted heteroaryl;
    • X2 is selected from the group consisting of Cl, OR24, N(R24)2 an optionally substituted C2-C6 alkyl, and M;
    • X'2 is selected from the group consisting of Cl, N(R24)2, and OR24;
    • Y2 and Y'2 are each independently O or NR24;
    • R24 is selected from the group of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
    • provided that at least one of R21 and X2 is M;
    • or a stereoisomer or a pharmaceutically acceptable salt thereof.
  • Some embodiments relate to a compound of Formula III:
    Figure imgb0009
    wherein:
    • R31 is H; or R31 optionally forms a bond with M or X3 when X3 is N;
    • R32 and R33 are each independently selected from the group consisting of Cl, OH, NH2, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, and an optionally substituted C1-C6 alkylamino;
    • M is a biological agent or part of a biological agent or a prodrug of a biological agent;
    • X3 is O or NR34;
    • Y3 is selected from the group consisting of O, NR34, and an optionally substituted C1-C6 alkyl;
    • R34 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
    • or a stereoisomer or a pharmaceutically acceptable salt thereof.
  • Some embodiments relate to a compound of Formula IV:
    Figure imgb0010
    wherein:
    • R41 is H; or R41 optionally forms a bond with M or X4 when X4 is N;
    • Z4 is selected from the group consisting of CR46R47, C(O), C(O)O, C(O)NR48, SO2, an optionally substituted aryl, and an optionally substituted heteroaryl;
    • R42, R43, R44, R45, R46, and R47 are each independently selected from the group consisting of H, OH, amino, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 acyloxy, an optionally substituted C1-C6 hereroalkyl, an optionally substituted phosphate, an optionally substituted phosphonate, an optionally substituted aryl, and an optionally substituted heterocycle; or R44 and R45 are independently or together optionally linked with R42, R43, R46, or R47 to form an optionally substituted ring; or R44 is optionally M; or R44 and R45 are together optionally to form an oxo (=O) or its derivative;
    • M is a biological agent or part of a biological agent or a prodrug of a biological agent;
    • X4 is selected from the group consisting of O, NR48, NC(O)R48, NS(O)2R49, and NP(O)(R50)2;
    • R48 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
    • R49 is selected from the group consisting of NH2, an optionally substituted C1-C6 alkyl, an optionally substituted aryl, and an optionally substituted heteroaryl;
    • R50 is selected from the group consisting of OH, NH2, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, and an optionally substituted C1-C6 alkylamino;
    • n is 0, 1, 2, or 3;
    • or a stereoisomer or a pharmaceutically acceptable salt thereof.
  • Some embodiments relate to a compound of Formula V:
    Figure imgb0011
    wherein:
    • R51 and R52 are each independently selected from the group consisting of H, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 acyloxy, an optionally substituted C1-C6 acylamino, an optionally substituted phosphate, an optionally substituted phosphonate, an optionally substituted phosphoramidate, an optionally substituted C1-C6 aryl, and an optionally substituted heteroaryl; or R51 and R52 are together optionally to form an oxo (=O) or its derivative;
    • R53 and R54 are each independently selected from the group consisting of Cl, OH, NH2, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 alkylamino, an optionally substituted C1-C6 acylamino, an optionally substituted aryloxy, an optionally substituted phosphate, an optionally substituted phosphonate, and an optionally substituted heteroaryloxy; or R53 is optionally linked with R51, R54, or R56 to form an optionally substituted 5-, 6-, or 7-membered heterocycle;
    • R55 is selected from the group consisting of OH, NH2, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 alkylamino, an optionally substituted aryl, and an optionally substituted heteroaryl; or R55 is optionally linked to R51 or R56 to form an optionally substituted ring;
    • R56 is selected from the group consisting of H, a C1-C6 alkyl, and a C1-C6 heteroalkyl;
    • M is a biological agent or part of a biological agent or a prodrug of a biological agent;
    • X5 is O or NR56;
    • n is 0, 1, 2, or 3;
    • or a stereoisomer or a pharmaceutically acceptable salt thereof.
  • Some embodiments relate to a compound of Formula VI:
    Figure imgb0012
    wherein:
    • R61 and R62 are each independently selected from the group consisting of H, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 alkylamino, an optionally substituted C1-C6 acyloxy, -OCH2P(O)(R69)2, and an optionally substituted C1-C6 acylamino; or R61 and R62 together optionally form an oxo (=O) or its derivative;
    • R63, R64, and R65 are each independently selected from the group consisting of H, CO2R67, C(O)N(R67)2, P(O)(R69)2, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 acyloxy, and an optionally substituted C1-C6 hereroalkyl; or two of R63, R64, and R65 are optionally linked to form an optionally substituted ring; or R63 is optionally linked with R68 to form an optionally substituted ring; with the proviso that CR63R64R65 is not a straight chain C1-C4 alkyl when R61 and R62 form an oxo (=O);
    • R66 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 heteroalkyl, an optionally substituted aryl, and an optionally substituted heteroaryl;
    • R67 and R68 are each independently selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
    • R69 is selected from the group consisting of OH, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, and an optionally substituted C1-C6 alkylamino;
    • M is a biological agent or part of a biological agent or a prodrug of a biological agent;
    • X6 is O or NR68;
    • Y6 is selected from the group consisting of null, O, NR68, and C(R68)2;
    • or a stereoisomer or a pharmaceutically acceptable salt thereof.
  • Some embodiments relate to a compound of Formula VII:
    Figure imgb0013
    wherein:
    • R71 is selected from the group consisting of H, OH, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted phosphate, and an optionally substituted phosphonate;
    • X7 is O or S;
    • Y7 is N or CR71;
    • M is a biological agent or part of a biological agent or a prodrug of a biological agent;
    • or a stereoisomer or a pharmaceutically acceptable salt thereof.
  • In some embodiments, the compound is selected from the group consisting of:
    Figure imgb0014
    Figure imgb0015
    Figure imgb0016
    Figure imgb0017
    and a stereoisomer or a pharmaceutically acceptable salt thereof.
  • In some embodiments, M is a nucleoside antiviral or anticancer agent.
  • In some embodiments, M is a lipid modulator.
  • In some embodiments, M is selected from the group consisting of HMG-CoA reductase inhibitor, a selective thyroid hormone receptor modulator, a peroxisome proliferator-activated receptor modulator, a fibrate, a nicotinic acid, a bile acid, and a fatty acid.
  • In some embodiments, M is a glucose modulator.
  • In some embodiments, M is selected from the group consisting of a peroxisome proliferator-activated receptor modulator, a glucose biosynthesis inhibitor, and a dipeptidyl peptidase 4 inhibitor.
  • In some embodiments, M is a nuclear hormone receptor modulator.
  • Some embodiments relate to a pharmaceutical composition comprising any of the above compounds and a pharmaceutically acceptable excipient.
  • Some embodiments relate to a method of treating a disease, disorder or condition comprising administering an effective amount of any of the above compounds to a subject in need thereof.
  • In some embodiments, the disease, disorder or condition is a disease, disorder or condition of the liver.
  • In some embodiments, the disease, disorder or condition is a metabolic, cardiovascular or hormonal disease in which the liver is involved in the production and/or the homeostasis control of the biochemical end products of the disease, disorder or condition.
  • In some embodiments, the disease, disorder or condition is selected from the group consisting of hepatitis, liver cancer, liver fibrosis, fatty liver, malaria, viral infection, parasitic infection, diabetes, hyperlipidemia, atherosclerosis, obesity, dyslipidemia, hyperglycemia and a hormonal condition.
  • Some embodiments relate to a method of treating a liver disease comprising administering an effective amount of any of the above compounds to a subject in need thereof, wherein M is a nucleoside antiviral or anticancer agent.
  • Some embodiments relate to a method of treating dyslipidemia comprising administering to a subject in need thereof an effective amount of any of the above compounds, wherein M is a lipid modulator.
  • In some embodiments, M is selected from the group consisting of HMG-CoA reductase inhibitor, a selective thyroid hormone receptor modulator, peroxisome proliferator-activated receptor modulator, a fibrate, nicotinic acid, a bile acid, and a fatty acid.
  • Some embodiments relate to method of treating hyperglycemia comprising administering to a subject in need thereof, an effective amount of any of the above compounds, wherein M is a glucose modulator.
  • In some embodiments, M is selected from the group consisting of peroxisome proliferator-activated receptor modulator, glucose biosynthesis inhibitor, and dipeptidyl peptidase 4 inhibitor.
  • Some embodiments relate to a method of treating a hormonal condition comprising administering to a subject in need thereof, an effective amount of any of the above compounds, wherein M is a nuclear hormone receptor modulator.
  • Some embodiments further comprise administering an effective amount of at least one additional therapeutic agent to the subject in need thereof.
  • Some embodiments relate to a method of delivering a diagnostic imaging agent to the liver of a subject in need thereof, comprising administering to the subject an effective amount of any of the above compounds.
  • In some embodiments, the subject is a mammal.
  • In some embodiments, the subject is human.
  • Some embodiments relate to a method of inhibiting viral replication in a cell comprising contacting the cell with any of the above compounds.
  • Some embodiments relate to a method of intervening in a molecular pathway or modulating a target in a cell comprising contacting the cell with any of the above compounds.
  • In some embodiments, the cell is in vivo.
  • In some embodiments, the cell is ex vivo.
  • In some embodiments, the cell is a hepatocyte.
  • In some embodiments, the cell is mammalian.
  • In some embodiments, the cell is human.
  • DETAILED DESCRIPTION
  • The present embodiments are directed to compositions and methods related to novel prodrug compounds of biologically active acid/alcohol derivatives such as phosphates, phosphonates, phosphonamidates, phosphoramidates, carboxylates, phenolates, and alkoxylates, their preparation and their uses. These prodrug compounds and their stereoisomers and pharmaceutically acceptable salts are represented by the formulae discussed below.
  • Some embodiments relate to a compound of Formula I:
    Figure imgb0018
    wherein:
    • R1 and R2 are each independently selected from the group consisting of H, M, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 acyloxy, an optionally substituted aryl, and an optionally substituted heteroaryl;
    • R3 is selected from the group consisting of H and an optionally substituted C1-C6 alkyl; or R3 optionally forms an optionally substituted ring with R2; or R3 together with R2 form a methylene or its derivative; or R3 together with R2 form an oxo (=O) or its derivative; or R3 optionally forms a bond with Z or Y' when Z or Y' is N;
    • M is a biological agent, or part of a biological agent or a prodrug of a biological agent;
    • X is selected from the group consisting of Cl, OR4, NR4R5, an optionally substituted C1-C6 alkyl, and M;
    • Y and Y' are each independently O or NR4; or Y' is CH2 or null;
    • Z is selected from the group consisting of O, NR5, CR8R6, C=O, C=NR7, and null; or Z is a 2-5 atom spacer selected from an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 heteroalkyl, an optionally substituted aryl, and an optionally substituted heteroaryl;
    • R4 is selected from the group of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
    • R5 is selected from the group consisting of H, M, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, and an optionally substituted C1-C6 acyl;
    • R6 is selected from the group consisting of F, Cl, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl; or R6 is H provided that R1 or R2 or R8 is connected to a cyclic core carbon atom through an oxygen-carbon (O-C) bond;
    • R7 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
    • R8 is selected from the group consisting of F, Cl, M, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl; or R8 is H provided that R1 or R2 is connected to a cyclic core carbon atom through an oxygen-carbon (O-C) bond;
    • provided that at least one of R1, R2, R5, R8, and X is M;
    • or a stereoisomer or a pharmaceutically acceptable salt thereof.
  • In some embodiments, the compound is a compound of Formula Ia:
    Figure imgb0019
    wherein:
    • R1 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, an optionally substituted aryl, and an optionally substituted heteroaryl;
    • R2 and R3 are each independently selected from the group consisting of H and an optionally substituted C1-C6 alkyl; or R2 and R3 optionally form an optionally substituted ring; or R2 and R3 optionally form a methylene or its derivative; or R2 and R3 optionally form an oxo or its derivative;
    • M is a biological agent, part of a biological agent or a prodrug of a biological agent;
    • Y and Y' are each independently O or NR4; or Y' is CH2;
    • Z is selected from the group consisting of O, NR5, CR8R6, C=O, C=NR7, and null; or Z is a 2-5 atom spacer selected from an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 heteroalkyl, an optionally substituted aryl, and an optionally substituted heteroaryl;
    • R4 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
    • R5 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, and an optionally substituted C1-C6 acyl;
    • R6 is selected from the group consisting of F, Cl, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl; or R6 is H provided that R1 or R2 or R8 is connected with the cyclic core carbon atom through an oxygen-carbon (O-C) bond;
    • R7 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
    • R8 is selected from the group consisting of F, Cl, M, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl; or R8 is H provided that R1 or R2 is connected to a cyclic core carbon atom through an oxygen-carbon (O-C) bond;
    • or a stereoisomer or a pharmaceutically acceptable salt thereof.
  • In some embodiments, the compound is a compound of Formula Ib:
    Figure imgb0020
    wherein:
    • R2 and R3 are independently selected from the group consisting of H and an optionally substituted C1-C6 alkyl; or R2 and R3 optionally form an optionally substituted ring; or R2 and R3 optionally form a methylene or its derivative; or R2 and R3 optionally form an oxo (=O) or its derivative;
    • M is a biological agent, part of a biological agent or a prodrug of a biological agent;
    • X is selected from the group consisting of Cl, an optionally substituted C1-C6 alkyl, NR4R5, and OR4;
    • Y and Y' are each independently O or NR4; or Y' is CH2 or null;
    • Z is selected from the group consisting of O, NR5, C(R6)2, C=O, C=NR7, and null; or Z is a 2-5 atom spacer selected from an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 heteroalkyl, an optionally substituted aryl, and an optionally substituted heteroaryl;
    • R4 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
    • R5 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, and an optionally substituted C1-C6 acyl;
    • R6 is selected from the group consisting of F, Cl, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl; or R6 is H provided that M or R2 is connected with the cyclic core carbon atom through an oxygen-carbon (O-C) bond;
    • R7 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
    • or a stereoisomer or a pharmaceutically acceptable salt thereof.
  • In some embodiments, the compound is a compound of Formula Ic:
    Figure imgb0021
    wherein:
    • R1 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, an optionally substituted aryl, and an optionally substituted heteroaryl;
    • R3 is selected from the group consisting of H and an optionally substituted C1-C6 alkyl; or R3 together with M form a methnylene derivative; or R3 optionally forms a bond with Z or Y' when Z or Y' is N;
    • M is a biological agent, part of a biological agent or a prodrug of a biological agent;
    • X is selected from the group consisting of Cl, OR4, NR4R5, and an optionally substituted C1-C6 alkyl;
    • Y and Y' are each independently O or NR4;
    • Z is selected from the group consisting of O, NR5, C(R6)2, C=O, C=NR7, and null; or Z is a 2-5 atom spacer selected from an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 heteroalkyl, an optionally substituted aryl, and an optionally substituted heteroaryl;
    • R4 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
    • R5 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, and an optionally substituted C1-C6 acyl;
    • R6 is selected from the group consisting of F, Cl, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl; or R6 is H provided that R1 or M is connected with the cyclic core carbon atom through an oxygen-carbon (O-C) bond;
    • R7 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
    • or a stereoisomer or a pharmaceutically acceptable salt thereof.
  • In some embodiments, the compound is a compound of Formula Id or Ie:
    Figure imgb0022
    wherein:
    • R2 and R3 are independently selected from the group consisting of H and an optionally substituted C1-C6 alkyl; or R3 together with M or R2 form a methylene derivative; or R3 together with M or R2 form an optionally substituted ring; or R3 together with R2 form an oxo (=O) or its derivative; or R3 optionally forms a bond with Z or Y' when Z or Y' is N;
    • M is a biological agent, part of a biological agent or a prodrug of a biological agent;
    • X is selected from the group consisting of Cl, OR4, and an optionally substituted C2-C6 alkyl;
    • Y and Y' are each independently O or N; or Y' is CH2;
    • Z is selected from the group consisting of O and NR5;
    • R4 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
    • R5 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, and an optionally substituted C1-C6 acyl;
    • or a stereoisomer or a pharmaceutically acceptable salt thereof.
  • Some embodiments relate to a compound of Formula IIa or IIb:
    Figure imgb0023
    Figure imgb0024
    wherein:
    • R21 is selected from the group consisting of H, M, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 acyloxy, an optionally substituted aryl, and an optionally substituted heteroaryl;
    • M is a biological agent, or part of a biological agent or a prodrug of a biological agent;
    • Q is an optionally substituted aryl or an optionally substituted heteroaryl;
    • X2 is selected from the group consisting of Cl, OR24, N(R24)2 an optionally substituted C2-C6 alkyl, and M;
    • X'2 is selected from the group consisting of Cl, N(R24)2, and OR24;
    • Y2 and Y'2 are each independently O or NR24;
    • R24 is selected from the group of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
    • provided that at least one of R21 and X2 is M;
    • or a stereoisomer or a pharmaceutically acceptable salt thereof.
  • Some embodiments relate to a compound of Formula III:
    Figure imgb0025
    wherein:
    • R31 is H; or R31 optionally forms a bond with M or X3 when X3 is N;
    • R32 and R33 are each independently selected from the group consisting of Cl, OH, NH2, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, and an optionally substituted C1-C6 alkylamino;
    • M is a biological agent or part of a biological agent or a prodrug of a biological agent;
    • X3 is O or NR34;
    • Y3 is selected from the group consisting of O, NR34, and an optionally substituted C1-C6 alkyl;
    • R34 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
    • or a stereoisomer or a pharmaceutically acceptable salt thereof.
  • Some embodiments relate to a compound of Formula IV:
    Figure imgb0026
    wherein:
    • R41 is H; or R41 optionally forms a bond with M or X4 when X4 is N;
    • Z4 is selected from the group consisting of CR46R47, C(O), C(O)O, C(O)NR48, SO2, an optionally substituted aryl, and an optionally substituted heteroaryl;
    • R42, R43, R44, R45, R46, and R47 are each independently selected from the group consisting of H, OH, amino, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 acyloxy, an optionally substituted C1-C6 hereroalkyl, an optionally substituted phosphate, an optionally substituted phosphonate, an optionally substituted aryl, and an optionally substituted heterocycle; or R44 and R45 are independently or together optionally linked with R42, R43, R46, or R47 to form an optionally substituted ring; or R44 is optionally M; or R44 and R45 are together optionally to form an oxo (=O) or its derivative;
    • M is a biological agent or part of a biological agent or a prodrug of a biological agent;
    • X4 is selected from the group consisting of O, NR48, NC(O)R48, NS(O)2R49, and NP(O)(R50)2;
    • R48 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
    • R49 is selected from the group consisting of NH2, an optionally substituted C1-C6 alkyl, an optionally substituted aryl, and an optionally substituted heteroaryl;
    • R50 is selected from the group consisting of OH, NH2, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, and an optionally substituted C1-C6 alkylamino;
    • n is 0, 1, 2, or 3;
    • or a stereoisomer or a pharmaceutically acceptable salt thereof.
  • Some embodiments relate to a compound of Formula Va, Vb, or Vc:
    Figure imgb0027
    wherein:
    • R51 and R52 are each independently selected from the group consisting of H, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 acyloxy, an optionally substituted C1-C6 acylamino, an optionally substituted phosphate, an optionally substituted phosphonate, an optionally substituted phosphoramidate, an optionally substituted C1-C6 aryl, and an optionally substituted heteroaryl; or R51 and R52 are together optionally to form an oxo (=O) or its derivative;
    • R53 and R54 are each independently selected from the group consisting of Cl, OH, NH2, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 alkylamino, an optionally substituted C1-C6 acylamino, an optionally substituted aryloxy, an optionally substituted phosphate, an optionally substituted phosphonate, and an optionally substituted heteroaryloxy; or R53 is optionally linked with R51, R54, or R56 to form an optionally substituted 5-, 6-, or 7-membered heterocycle;
    • R55 is selected from the group consisting of OH, NH2, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 alkylamino, an optionally substituted aryl, and an optionally substituted heteroaryl; or R55 is optionally linked to R51 or R56 to form an optionally substituted ring;
    • R56 is selected from the group consisting of H, a C1-C6 alkyl, and a C1-C6 heteroalkyl;
    • M is a biological agent or part of a biological agent or a prodrug of a biological agent;
    • X5 is O or NR56;
    • n is 0, 1, 2, or 3;
    • or a stereoisomer or a pharmaceutically acceptable salt thereof.
  • Some embodiments relate to a compound of Formula VIa, VIb or VIc:
    Figure imgb0028
    wherein:
    • R61 and R62 are each independently selected from the group consisting of H, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 alkylamino, an optionally substituted C1-C6 acyloxy, -OCH2P(O)(R69)2, and an optionally substituted C1-C6 acylamino; or R61 and R62 together optionally form an oxo (=O) or its derivative;
    • R63, R64, and R65 are each independently selected from the group consisting of H, CO2R67, C(O)N(R67)2, P(O)(R69)2, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 acyloxy, and an optionally substituted C1-C6 hereroalkyl; or two of R63, R64, and R65 are optionally linked to form an optionally substituted ring; or R63 is optionally linked with R68 to form an optionally substituted ring; with the proviso that CR63R64R65 is not a straight chain C1-C4 alkyl when R61 and R62 form an oxo (=O);
    • R66 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 heteroalkyl, an optionally substituted aryl, and an optionally substituted heteroaryl;
    • R67 and R68 are each independently selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
    • R69 is selected from the group consisting of OH, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, and an optionally substituted C1-C6 alkylamino;
    • M is a biological agent or part of a biological agent or a prodrug of a biological agent;
    • X6 is O or NR68;
    • Y6 is selected from the group consisting of null, O, NR68, and C(R68)2;
    • or a stereoisomer or a pharmaceutically acceptable salt thereof.
  • Some embodiments relate to a compound of Formula VII:
    Figure imgb0029
    wherein:
    • R71 is selected from the group consisting of H, OH, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted phosphate, and an optionally substituted phosphonate;
    • X7 is O or S;
    • Y7 is N or CR71;
    • M is a biological agent or part of a biological agent or a prodrug of a biological agent;
    • or a stereoisomer or a pharmaceutically acceptable salt thereof.
  • CYP3A4 is expressed in the liver in a level much higher than other tissues (DeWaziers et al. J Pharm Exp Ther 253:387 (1990)). Prodrug compounds of Formula I-VII are predominantly activated via CYP3A4 in the liver. In some embodiments, the prodrug compounds of Formulae I-VII have high efficiency in liver-targeting via selective delivery of biologically active agents to the liver. In some embodiments, the prodrugs are used to increase the therapeutic index of the drug, since the prodrug compounds of Formulae I-VII may not be active or may be less active outside the liver.
  • Certain drugs of phosph(on)ate derivatives are highly charged compounds that have generally poor oral bioavailability due to poor absorption in the gastrointestinal tract. Certain drugs are highly lipophilic compounds that have generally poor oral bioavailability due to poor absorption in the gastrointestinal tract. In some embodiments, the prodrug compounds of Formulae I-VII have oral bioavailability superior to the parent drugs/agents.
  • In some embodiments, due to the liver-targeting nature of the prodrug compounds of Formulae I-VII, the compounds are used to treat diseases that benefit from enhanced drug distribution to the liver and like tissues and cells, including but not limited to diseases in the liver, such as hepatitis, liver cancer, liver fibrosis, fatty liver, malaria, other viral and parasitic infections, and metabolic, cardiovascular, and/or hormonal diseases where the liver is involved in the production and/or the homeostasis control of the biochemical end products, e.g. glucose (diabetes); cholesterol, fatty acids, bile acids, triglycerides (hyperlipidemia) (atherosclerosis) (obesity), lipoproteins, apolipoproteins, and sex hormone-binding globulin (SHBG).
  • In some embodiments, the disclosed prodrugs are used to prolong pharmacodynamic half-life of the drug. In addition, the disclosed prodrug methodology can be used to achieve sustained delivery of the parent drug. In some embodiments, a method of making these prodrugs is described. In some embodiments, the prodrugs are also useful in the delivery of diagnostic imaging agents to the liver or other tissues.
  • Some compounds of Formulae I-VII have asymmetric centers where the stereochemistry is unspecified, and the diastereomeric mixtures of these compounds are included, as well as the individual stereoisomers when referring to a compound of Formulae I-VII generally.
  • Some embodiments of the compounds, compositions and methods provided herein include a pharmaceutical composition comprising a compound provided herein and a pharmaceutically acceptable carrier.
  • Some embodiments also include administering an effective amount of a second or multiple therapeutic agents in combination with a compound provided herein to the subject in need thereof.
  • Some embodiments of the compounds, compositions and methods provided herein include a method of treating a liver disease comprising administering an effective amount of a compound provided herein where M is a nucleoside antiviral or anticancer agent to a subject in need thereof.
  • Some embodiments of the compounds, compositions and methods provided herein include a method of treating dyslipidemia comprising administering an effective amount of a compound provided herein where M is a lipid modulator such as a HMG-CoA reductase inhibitor, a selective thyroid hormone receptor modulator, peroxisome proliferator-activated receptor modulator, a fibrate, nicotinic acid, and an omega-3 fatty acid to a subject in need thereof.
  • Some embodiments of the compounds, compositions and methods provided herein include a method of treating hyperglycemia comprising administering an effective amount of a compound provided herein where M is a glucose modulator such as peroxisome proliferator-activated receptor modulator, glucose biosynthesis inhibitor, and dipeptidyl peptidase 4 inhibitor to a subject in need thereof.
  • Some embodiments of the compounds, compositions and methods provided herein include a method of treating a hormonal condition comprising administering an effective amount of a compound provided herein where M is a nuclear hormone receptor modulator to a subject in need thereof.
  • In some embodiments, the subject is mammalian.
  • In some embodiments, the subject is human.
  • Some embodiments of the compounds, compositions and methods provided herein include a method of testing a compound in a cell comprising contacting the cell with the compound of the claims.
  • In some embodiments, the cell is in vivo.
  • In some embodiments, the cell is ex vivo.
  • In some embodiments, the cell is a hepatocyte.
  • In some embodiments, the cell is mammalian.
  • In some embodiments, the cell is human.
  • Some embodiments of the compounds, compositions and methods provided herein include use of a compound provided herein in the treatment of a disease in the liver.
  • Some embodiments include the use of a compound provided herein in combination with additional therapeutic agent(s) for the treatment of a disease in the liver.
  • Some embodiments of the compounds, compositions and methods provided herein include use of a compound provided herein in the treatment of a disease or condition by intervening in a molecular pathway in the liver.
  • Some embodiments include the use of a compound provided herein in combination with additional therapeutic agent(s) for the treatment of a disease or condition by intervening in a molecular pathway in the liver.
  • Definitions
  • In accordance with the present disclosure and as used herein, the following terms are defined with the following meanings, unless explicitly stated otherwise. It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the subject matter claimed. In this application, the use of the singular includes the plural unless specifically stated otherwise. In this application, the use of "or" means "and/or" unless stated otherwise. Furthermore, use of the term "including" as well as other forms, such as "includes," and "included" is not limiting.
  • As used herein, ranges and amounts can be expressed as "about" a particular value or range. "About" also includes the exact amount. Hence "about 10%" means "about 10%" and also "10%."
  • As used herein, "optional" or "optionally" means that the subsequently described event or circumstance does or does not occur, and that the description includes instances where the event or circumstance occurs and instances where it does not. For example, an optionally substituted group means that the group is unsubstituted or is substituted.
  • As used herein, the singular forms "a," "an" and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to a composition comprising "a therapeutic agent" includes compositions with one or a plurality of therapeutic agents.
  • The term "alkyl" refers to saturated aliphatic groups including straight-chain, branched chain and cyclic groups, up to and including 10 carbon atoms. Suitable alkyl groups include methyl, ethyl, n-propyl, isopropyl, and cyclopropyl. The alkyl group may be optionally substituted with 1-3 substituents.
  • The term "optionally substituted" or "substituted" includes groups substituted by one to four substituents, independently selected from lower alkyl, lower aryl, lower aralkyl, lower cyclic alkyl, lower heterocycloalkyl, hydroxy, lower alkoxy, lower aryloxy, perhaloalkoxy, aralkoxy, lower heteroaryl, lower heteroaryloxy, lower heteroarylalkyl, lower heteroaralkoxy, azido, amino, halogen, lower alkylthio, oxo, lower acylalkyl, lower carboxy esters, carboxyl, carboxamido, nitro, lower acyloxy, lower aminoalkyl, lower alkylaminoaryl, lower alkylaryl, lower alkylaminoalkyl, lower alkoxyaryl, lower arylamino, lower aralkylamino, lower alkylsulfonyl, lower carboxamidoalkylaryl, lower carboxamidoaryl, lower hydroxyalkyl, lower haloalkyl, lower alkylaminoalkylcarboxy, lower aminocarboxamidoalkyl, cyano, lower alkoxyalkyl, lower perhaloalkyl, phosphate, phosphonate, or phosphoramidate, and lower arylalkyloxyalkyl. "Substituted aryl" and "substituted heteroaryl" refers to aryl and heteroaryl groups substituted with 1-6 substituents. These substituents are selected from the group consisting of lower alkyl, lower alkoxy, lower perhaloalkyl, halogen, hydroxy, cyano, and amino.
  • The term "heteroalkyl" refer to alkyl groups containing at least one heteroatom, such as 1 to 3 heteroatoms. Suitable heteroatoms include oxygen, sulfur, and nitrogen.
  • The term "heteroacyl" refer to -C(O)-heteroalkyl groups.
  • The term "acyloxy" refers to -OC(O)R where R is alkyl, or heteroalkyl.
  • The term "alkoxy" or "alkyloxy" refers to OR where R is alkyl, or heteroalkyl, all optionally substituted.
  • The term "carboxyl" refers to C(O)OH.
  • The term "oxo" refers to an =O group.
  • The term "oxo derivative" refers to =NR where R is H, lower alkyl, lower alkoxyl, or lower alkylamino.
  • The term "amino" refers to NRR' where R and R' are each independently selected from hydrogen, alkyl, aryl, aralkyl and heterocycloalkyl, all except H are optionally substituted; and R and R' can form a cyclic ring system.
  • The term 'acylamino" refers to -NRC(O)R' where R and R' are each independently selected from H, alkyl, or heteroalkyl.
  • The term "halogen" or "halo" refers to F, Cl, Br and I.
  • The term "haloalkyl" refer to alkyl groups containing at least one halogen, in a further aspect are 1 to 3 haloatoms. Suitable haloatoms include F, Cl, and Br.
  • The term "haloheteroalkyl" refer to alkyl groups containing at least one halogen and one heteroatom.
  • The term "haloacyl" refer to -C(O)-haloalkyl groups.
  • The term "haloheteroacyl" refer to -C(O)-haloheteroalkyl groups.
  • The term "alkenyl" refers to unsaturated groups which have 2 to 12 atoms and contain at least one carbon carbon double bond and includes straight chain, branched chain and cyclic groups. Alkenyl groups may be optionally substituted. Suitable alkenyl groups include allyl.
  • The term "alkynyl" refers to unsaturated groups which have 2 to 12 atoms and contain at least one carbon carbon triple bond and includes straight chain, branched chain and cyclic groups. Alkynyl groups may be optionally substituted. Suitable alkynyl groups include ethynyl.
  • The term "methylene" refers to =CH2.
  • The term "methylene derivative" refers to =CRR' where R and R' are each independently selected from an optionally substituted alkyl, an optionally substituted alkenyl, and M.
  • The term "aminoalkyl" refers to the group NR2-alkyl where R is selected from H, alkyl, aryl, aralkyl, and heterocycloalkyl.
  • The terms "alkylthio" refers to the group alkyl-S-.
  • The term "amido" refers to the NR2 group next to an acyl or sulfonyl group as in NR2C(O)-, RC(O)NR-, NR2S(=O)2- and RS(=O)2-NR-, where R includes H, alkyl, aryl, aralkyl, and heterocycloalkyl.
  • The term "perhalo" refers to groups wherein every C-H bond has been replaced with a C-halo bond on an aliphatic or aryl group. Suitable perhaloalkyl groups include CF3 and CFCl2.
  • The term "aryl" refers to an aromatic group wherein each of the atoms forming the ring is a carbon atom. Aryl rings may be formed by five, six, seven, eight, nine, or more than nine carbon atoms. Aryl groups may be optionally substituted. Examples of aryl groups include, but are not limited to phenyl, naphthalenyl, phenanthrenyl, anthracenyl, tetralinyl, fluorenyl, indenyl, and indanyl.
  • The term "heteroaryl" refers to an aromatic group wherein at least one atom forming the aromatic ring is a heteroatom. Heteroaryl rings may be formed by three, four, five, six, seven, eight, nine, or more than nine atoms. Heteroaryl groups may be optionally substituted. Examples of heteroaryl groups include, but are not limited to, aromatic C3-8 heterocyclic groups comprising one oxygen or sulfur atom or up to four nitrogen atoms, or a combination of one oxygen or sulfur atom and up to two nitrogen atoms, and their substituted as well as benzo- and pyrido-fused derivatives, for example, connected via one of the ring-forming carbon atoms. In some embodiments, heteroaryl groups are optionally substituted with one or more substituents, independently selected from halo, hydroxy, amino, cyano, nitro, alkylamido, acyl, C1-6-alkoxy, C1-6-alkyl, C1-6-hydroxyalkyl, C1-6-aminoalkyl, C1-6-alkylamino, alkylsulfenyl, alkylsulfinyl, alkylsulfonyl, sulfamoyl, or trifluoromethyl. Examples of heteroaryl groups include, but are not limited to, unsubstituted and mono- or di-substituted derivatives of furan, benzofuran, thiophene, benzothiophene, pyrrole, pyridine, indole, oxazole, benzoxazole, isoxazole, benzisoxazole, thiazole, benzothiazole, isothiazole, imidazole, benzimidazole, pyrazole, indazole, tetrazole, quinoline, isoquinoline, pyridazine, pyrimidine, purine and pyrazine, furazan, 1,2,3-oxadiazole, 1,2,3-thiadiazole, 1,2,4-thiadiazole, triazole, benzotriazole, pteridine, phenoxazole, oxadiazole, benzopyrazole, quinolizine, cinnoline, phthalazine, quinazoline, and quinoxaline. In some embodiments, the substituents are halo, hydroxy, cyano, O-C1-6-alkyl, C1-6-alkyl, hydroxy-C1-6-alkyl, and amino-C1-6-alkyl.
  • The term "aryloxy" refers to -O-aryl.
  • The term "heteroaryloxy" refers to -O-heteroaryl.
  • The phrase "therapeutically effective amount" means an amount of a compound or a combination of compounds that partially or fully ameliorates, attenuates or eliminates one or more of the symptoms of a particular disease or condition or prevents, modifies, or delays the onset of one or more of the symptoms of a particular disease or condition. Such amount can be administered as a single dosage or can be administered according to a regimen, whereby it is effective. Repeated administration may be needed to achieve a desired result (e.g., treatment of the disease and/or condition).
  • The term "pharmaceutically acceptable salt" includes salts of compounds of Formulae I-VII and its prodrugs derived from the combination of a compound of the present embodiments and an organic or inorganic acid or base. Suitable acids include acetic acid, adipic acid, benzenesulfonic acid, (+)-7,7-dimethyl-2-oxobicyclo[2.2.1]heptane-1-methanesulfonic acid, citric acid, 1,2-ethanedisulfonic acid, dodecyl sulfonic acid, fumaric acid, glucoheptonic acid, gluconic acid, glucuronic acid, hippuric acid, hydrochloride hemiethanolic acid, HBr, HCl, HI, 2-hydroxyethanesulfonic acid, lactic acid, lactobionic acid, maleic acid, methanesulfonic acid, methylbromide acid, methyl sulfuric acid, 2-naphthalenesulfonic acid, nitric acid, oleic acid, 4,4'-methylenebis-[3-hydroxy-2-naphthalenecarboxylic acid], phosphoric acid, polygalacturonic acid, stearic acid, succinic acid, sulfuric acid, sulfosalicylic acid, tannic acid, tartaric acid, terphthalic acid, and p-toluenesulfonic acid. Suitable bases include NaOH, KOH, Ca(OH)2, Zn(OH)2, Mg(OH)2, diethylamine, ethanolamine, diethanolamine, choline, lysine, meglumine, benzathine, and tromethamine.
  • The term "spacer" refers to an atom or group of atoms that separate two or more groups from one another by a desired number of atoms. For example, in some embodiments, it may be desirable to separate two or more groups by one, two, three, four, five, six, or more than six atoms. In such embodiments, any atom or group of atoms may be used to separate those groups by the desired number of atoms. Spacers are optionally substituted. In some embodiments, a spacer comprises saturated or unsaturated alkyls, heteroalkyls and/or haloalkyls. In some embodiments, a spacer comprises atoms that are part of a ring.
  • Where the number of any given substituent is not specified (e.g., "haloalkyl"), there may be one or more substituents present. For example, "haloalkyl" can include one or more of the same or different halogens. For example, "haloalkyl" includes each of the substituents CF3, CHF2 and CH2F.
  • Solely for the purposes of illustration, and without limiting the above definitions, some examples of spacers are provided. Examples of 2 atom spacers include, but are not limited to, the following:
    Figure imgb0030
    where A and B represent groups which are separated by the desired number of atoms.
  • Examples of 3 atom spacers include, but are not limited to, the following:
    Figure imgb0031
    Figure imgb0032
    where A and B represent groups which are separated by the desired number of atoms.
  • The term "patient" refers to an animal being treated including a mammal, such as a dog, a cat, a cow, a horse, a sheep, and a human. In some embodiments the patient is a mammal, either male or female. In some embodiments, the patient is a male or female human.
  • The term "prodrug" as used herein refers to any compound that when administered to a biological system generates a biologically active compound as a result of spontaneous chemical reaction(s), enzyme catalyzed chemical reaction(s), and/or metabolic chemical reaction(s), or a combination of each. Standard prodrugs are formed using groups attached to functionality, e.g. HO-, HS-, HOOC-, HOOPR2-, associated with the drug, that cleave in vivo. Standard prodrugs include but are not limited to carboxylate esters where the group is alkyl, aryl, aralkyl, acyloxyalkyl, alkoxycarbonyloxyalkyl as well as esters of hydroxyl, thiol and amines where the group attached is an acyl group, an alkoxycarbonyl, aminocarbonyl, phosphate or sulfate. The groups illustrated are examples, not exhaustive, and one skilled in the art could prepare other known varieties of prodrugs. Such prodrugs of the compounds of Formula I fall within this scope. Prodrugs must undergo some form of a chemical transformation to produce the compound that is biologically active or is a precursor of the biologically active compound. In some cases, the prodrug is biologically active, usually less than the drug itself, and serves to improve drug efficacy or safety through improved oral bioavailability, pharmacodynamic half-life, etc. Prodrug forms of compounds may be utilized, for example, to improve bioavailability, improve subject acceptability such as by masking or reducing unpleasant characteristics such as bitter taste or gastrointestinal irritability, alter solubility such as for intravenous use, provide for prolonged or sustained release or delivery, improve ease of formulation, or provide site specific delivery of the compound.
  • The term "stereoisomer" refers to the relative or absolute spatial relationship of the R group(s) and the substituent attached to the phosphorus atom via an exocyclic single bond on the 2-oxo-phosphorus prodrug ring, and refers to individual or any combination of the individual isomers such as a racemic mixture and a diastereomeric mixture. When one R group is attached to the carbon atom in the ring, there are four stereoisomers. For example, the structures A, B, C, and D below show four possible individual isomers. Structures A and D (or B and C) are a pair of two enantiomers (or called optical isomers). When two R groups attached to two different carbon atoms in the ring, there are eight possible stereoisomers.
    Figure imgb0033
  • The term "liver" refers to the liver organ.
  • The term "liver specificity" refers to the ratio: drug or a drug metabolite in liver tissue drug or a drug metabolite in blood or another tissue
    Figure imgb0034
  • as measured in animals treated with the drug or a prodrug. The ratio can be determined by measuring tissue levels at a specific time or may represent an AUC (area under a curve) based on values measured at three or more time points.
  • The term "increased or enhanced liver specificity" refers to an increase in liver specificity ratio in animals treated with the prodrug relative to animals treated with the parent drug. Compounds disclosed in US Patent No. 8,063,025 , US Patent No. 7,666,855 , and PCT Pub. No. WO2009/073506 , are designed for the liver-specific delivery of nucleosides for the treatment of HCV patients and take advantage of a cytochrome P450 enzyme that is mainly expressed in the liver.
  • The term "enhanced oral bioavailability" refers to an increase of at least about 50% of the absorption of the dose of the parent drug. In an additional aspect, the increase in oral bioavailability of the prodrug (compared to the parent drug) is at least about 100%, or a doubling of the absorption. Measurement of oral bioavailability usually refers to measurements of the prodrug, drug, or drug metabolite in blood, plasma, tissues, or urine following oral administration compared to measurements following parenteral administration.
  • The term "therapeutic index" refers to the ratio of the dose of a drug or prodrug that produces a therapeutically beneficial response relative to the dose that produces an undesired response such as death, an elevation of markers that are indicative of toxicity, and/or pharmacological side effects.
  • The term "sustained delivery" refers to an increase in the period in which there is a prolongation of therapeutically-effective drug levels due to the presence of the prodrug.
  • The terms "treating" or "treatment" of a disease includes inhibiting the disease (slowing or arresting or partially arresting its development), providing relief from the symptoms or side effects of the disease (including palliative treatment), and/or relieving the disease (causing regression of the disease).
  • The terms "biological agent" refers to a compound that has biological activity or that has molecular properties that can be used for diagnosis purpose, such as a compound carrying a radioactive isotope or a heavy atom.
  • The terms "molecular pathway" refers to a series of molecular events in tissues such as a receptor modulating sequence or a biosynthesis sequence that is involved in physiological or pathophysiological functions of a living animal.
  • The terms "vinyl ketone reactive intermediate" refers to compounds of the structure below that are chemically reactive to generate a covalent bond with a molecular entity in the tissues or cell, where R is H, alkyl, aryl, or heteroaryl.
    Figure imgb0035
  • Formulations
  • The disclosed compounds may be used alone or in combination with other treatments. These compounds, when used in combination with other agents, may be administered as a daily dose or an appropriate fraction of the daily dose (e.g., bid). The compounds may be administered after a course of treatment by another agent, during a course of therapy with another agent, administered as part of a therapeutic regimen, or may be administered prior to therapy by another agent in a treatment program.
  • Examples of pharmaceutically acceptable salts include acetate, adipate, besylate, bromide, camsylate, chloride, citrate, edisylate, estolate, fumarate, gluceptate, gluconate, glucuronate, hippurate, hyclate, hydrobromide, hydrochloride, iodide, isethionate, lactate, lactobionate, maleate, mesylate, methylbromide, methylsulfate, napsylate, nitrate, oleate, palmoate, phosphate, polygalacturonate, stearate, succinate, sulfate, sulfosalicylate, tannate, tartrate, terphthalate, tosylate, triethiodide, sodium, potassium, calcium, zinc, magnesium, diethylamine, ethanolamine, diethanolamine, cholinate, lysine, meglumine, benzathine, and tromethamine.
  • Compositions containing the active ingredient may be in any form suitable for the intended method of administration. In some embodiments, the compounds of a method and/or composition described herein can be provided via oral administration, rectal administration, transmucosal administration, intestinal administration, enteral administration, topical administration, transdermal administration, intrathecal administration, intraventricular administration, intraperitoneal administration, intranasal administration, intraocular administration and/or parenteral administration.
  • When the compounds are administered via oral administration, for example, tablets, troches, lozenges, aqueous or oil suspensions, dispersible powders or granules, emulsions, hard or soft capsules, syrups or elixirs may be prepared. Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation. Tablets containing the active ingredient in admixture with non-toxic pharmaceutically acceptable excipient which are suitable for manufacture of tablets are acceptable. These excipients may be, for example, inert diluents, such as calcium or sodium carbonate, lactose, calcium or sodium phosphate; granulating and disintegrating agents, such as maize starch, or alginic acid; binding agents, such as starch, gelatin or acacia; and lubricating agents, such as magnesium stearate, stearic acid or talc. Tablets may be uncoated or may be coated by known techniques including microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax may be employed.
  • Formulations for oral use may be also presented as hard gelatin capsules where the active ingredient can be mixed with an inert solid diluent, for example calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient can be mixed with water or an oil medium, such as peanut oil, liquid paraffin or olive oil.
  • Formulations suitable for parenteral administration include aqueous and non-aqueous isotonic sterile injection solutions which may contain, for example, antioxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The formulations may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
  • In some embodiments unit dosage formulations contain a daily dose or unit, daily sub-dose, or an appropriate fraction thereof, of a drug. It will be understood, however, that the specific dose level for any particular patient will depend on a variety of factors including the activity of the specific compound employed; the age, body weight, general health, sex and diet of the individual being treated; the time and route of administration; the rate of excretion; other drugs which have previously been administered; and the severity of the particular disease undergoing therapy, as is well understood by those skilled in the art.
  • Synthesis of compounds
  • The following procedures for the preparation of the cyclic prodrug compounds illustrate the general procedures used to prepare the compounds which apply to phosphate, phosphonate, phosphoramidate, carboxylic acid, and alcohol containing drugs. Prodrugs can be introduced at different stages of synthesis of a drug. In some embodiments, they are made at a later stage, because of the general sensitivity of these groups to various reaction conditions. Optically pure prodrugs containing a single isomer at the phosphorus center can be made, for example, by separation of the diastereomers by a combination of column chromatography and/or crytallyzation, or by enantioselective synthesis of chiral activated phosph(on)ate intermediates.
  • Scheme I describes general strategies of synthesis of the cyclic phosph(on)ate prodrug compounds of Formula I. The first strategy starts with treatment of a dihydroxyl compound of structure 1 where Z is not a heteroatom with phosphorus oxychloride to generate a cyclic phosphate of structure 2. A coupling reaction of a cyclic phosphate chloride of structure 2 with an alcohol derivative compound of structure 3 provides the final compound of structure 4. When Z is a heteroatom, an alternative strategy can be used where the alcohol compound of structure 3 is converted to its monophosphate of structure 5 via a two-step sequence of phosphorylation with phosphorus oxychloride and acid hydrolysis by a resin. The monophosphate of structure 5 is then treated with a dichloro compound of structure 6 in the presence of silver nitrate to give the final product of structure 4.
    Figure imgb0036
  • Scheme II describes general strategies of synthesis of cyclic phosphoramidate or phosphonamidate prodrug compounds of Formula I. Treatment of the hydroxylamino compounds of structure 7 (where Z is not a heteroatom) with phosphorus oxychloride provides the cyclic phosphoramidate of structure 8. A biological agent having a hydroxyl group of structure 3 is coupled with a prodrug intermediate of structure 8 to yield the final product of structure 9. Alternatively, a chloroamino compound of structure 10 (where Z is a heteroatom) is treated with phosphorus oxychloride and then an ion-exchange resin sequentially to give the intermediate of structure 11 that undergoes an internal cyclization followed by chlorination to the cyclic phosphoramidate of structure 8.
    Figure imgb0037
  • Scheme III describes general synthetic procedures of the 2-oxodioxazaphinine prodrugs of Formula I. A biological agent derivative of structure 12 is treated with phosphorus oxychloride to provide an intermediate of structure 13 that is then treated with a base in the presence of an aldehyde of structure 14 to give final product of structure 15. The regioisomers of structure 15 are made from a different route where a compound of structure 16 is treated sequentially with diphenyl phosphate under basic a condition to give an intermediate of structure 17. Base mediated internal cyclization of the intermediate of structure 17 affords the final product of structure 18.
    Figure imgb0038
  • Scheme IV describes general synthetic procedures of the prodrugs of Formula IIa. Benzaldehyde derivative of structure 19 is treated with phosphorus oxychloride to provide an intermediate of structure 20 that is then converted to structure 21 in the presence of a acid chloride derivative of a biologic agent with zinc chloride as the catalyst. Deprotection followed by treatment of a base affords the final product of structure 22.
    Figure imgb0039
  • Scheme V describes a procedure for preparing cyclic bisphosphonate prodrug compounds of Formula III via either an acid catalyzed condensation or a base catalyzed alkylation routes. Condensation reaction of a bisphosphonate of structure 23 with an aldehyde of structure 24 in the presence of an acid catalyst affords a product of structure 25. Alternatively, alkylation of a bisphosphonate of structure 23 with a halide compound of structure 26 in the presence of silver nitrate and/or a base provides the same product of structure 25.
    Figure imgb0040
  • Scheme VI describes general strategies of synthesis of the cyclic acetal prodrug compounds of Formula IV. The dihydroxyl compound of structure 1 is condensed with an aldehyde of structure 2 in the presence of catalytic amount of acid to give a product of structure 3. Aldehyde compound of structure 2 is prepared from the corresponding carboxylic acid by the standard procedure in the literature.
    Figure imgb0041
  • Compounds of Formula V-VII are prepared by using standard conditions in the literature from the corresponding acids and derivatives (e.g., See J. E. Starrett, Jr., et al. J Med Chem 37:1857-1864 (1994) and J. K. Dickson, et al. J Med Chem 39:661-664 (1996)).
  • EXAMPLES
  • It will be understood that the following are examples and that the present embodiments are not limited to these examples.
  • Some biologically active compounds of Formulae I-VII are prepared as outlined below. Some biologically nonactive compounds of Formulae I-VII are also prepared to demonstrate the synthetic methodologies as outlined below.
  • Example 1 (2R,3R,4R,5R)-2-(((7-Chloro-3-oxido-1,5-dihydrobenzo[e][1,3,2]dioxaphosphepin-3-yl)oxy)methyl)-5-(2,4-dioxo-3,4-dihydropyrimidin-1(2H)-yl)-4-fluoro-4-methyltetrahydrofuran-3-yl propionate (Compound 101)
  • Figure imgb0042
  • Compound 101 was prepared as a mixture of two diastereomers according to synthetic strategy of Scheme I from (4-chloro-1,2-phenylene)dimethanol and the nucleoside derivative. [M+H]+ calcd for C21H23ClFN2O9P: 533.08; found: 533.1. 1H NMR (400MHz, DMSO-d6 ) 11.60 (s, 1H), 7.74 (d, J = 8.4, 1H), 7.66 (dd, J = 4.8 and 1.6, 1H), 7.56-7.52 (m, 2H), 6.08 (bs, 1H), 5.72 (d, J = 8.4, 1H), 5.40-5.10 (m, 4H), 4.45-4.31 (m, 3H), 3.41-3.31 (m, 1H), 2.46 (q, J = 7.6, 2H), 1.33 (d, J = 22.8, 3H), and 1.06 (t, J = 7.6, 3H).
  • Example 2 (2R,3R,4R,5R)-2-(((4-(3-Chlorophenyl)-2-oxido-1,3,5,2-trioxaphosphinan-2-yl)oxy)methyl)-5-(2,4-dioxo-3,4-dihydropyrimidin-1(2H)-yl)-4-fluoro-4-methyltetrahydrofuran-3-yl acetate (Compound 102)
  • Figure imgb0043
  • Compound 102 can be prepared according to synthetic strategy of Scheme I from 3-chlorobenzaldehyde and the nucleoside derivative. MH+ = 534.06(Calc.).
  • Example 3 (2R,3R,4R,5R)-2-(((6-(3-Chlorophenyl)-5,5-dimethyl-2-oxido-1,3,2-oxazaphosphinan-2-yl)oxy)methyl)-5-(2,4-dioxo-3,4-dihydropyrimidin-1(2H)-yl)-4-fluoro-4-methyltetrahydrofuran-3-yl acetate (Compound 103)
  • Figure imgb0044
  • Compound 103 was prepared as a 1:1 mixture of two diastereomers according to synthetic strategy of Scheme II from 3-amino-1-(3-chlorophenyl)-2,2-dimethylpropan-1-ol and the nucleoside derivative. [M+H]+ calcd for C23H28ClFN3O8P: 561.13; found: 561.2. 1H NMR (400MHz, DMSO-d6 ) 11.60 (s, 0.5H), 11.50 (s, 0.5H), 7.80-7.20 (m, 5H), 6.02 (bs, 1H), 5.55-5.50 (m, 1H), 5.40 (s, 1H), 4.38-4.02 (m, 5H), 2.13 (s, 3H), 1.34 (d, J = 15.5, 1.5H), 1.28 (d, J = 15.5, 1.5H), 0.92 (s, 3H, and 0.73 (s, 3H).
  • Example 4 (2R,3R,4R,5R)-2-(((5-(3-Chlorophenyl)-1-isopropyl-2-oxido-1,3,2-oxazaphospholidin-2-yl)oxy)methyl)-5-(2,4-dioxo-3,4-dihydropyrimidin-1(2H)-yl)-4-fluoro-4-methyltetrahydrofuran-3-yl acetate (Compound 104)
  • Figure imgb0045
  • Compound 104 was prepared as a mixture of two diastereomers according to synthetic strategy of Scheme II from 2-isopropylamino-1-(3-chlorophenyl)ethan-1-ol and the nucleoside derivative. [M+H]+ calcd for C23H28ClFN3O8P: 560.13; found: 560.2. 1H NMR (400MHz, DMSO-d6 ) 11.59 (s, 1H), 7.70-7.60 (m, 1H), 7.50 (d, J = 9.6, 1H), 7.44-7.40 (m, 3H), 6.10 (bs, 1H), 5.59-5.56 (m, 1H), 5.45 (bs, 1H), 5.22 (bs, 1H), 4.40-4.16 (m, 3H), 3.90-3.72 (m, 1H), 3.40 9bs, 1H), 3.14 (bs, 1H), 2.14 (s, 3H), and 1.38-1.12 (m, 9H).
  • Example 5 2-Ethoxy-4,6-di(pyridine-3-yl)-1,5,3,2-dioxazaphosphinine 2-oxide (Compound 105)
  • Figure imgb0046
  • Compound 105 can be prepared according to synthetic strategy of Scheme II from nicotinic acid and pyridine-3-aldehyde. MH+ = 319.07(Calc.).
  • Example 6 (2R,3R,4R,5R)-2-(((7-(3-chlorophenyl)-4-methyl-2-oxido-1,3,4,2-dioxazaphosphepan-2-yl)oxy)methyl)-5-(2,4-dioxo-3,4-dihydropyrimidin-1(2H)-yl)-4-fluoro-4-methyltetrahydrofuran-3-yl acetate (Compound 106)
  • Figure imgb0047
  • Compound 106 was prepared as a mixture of two diastereomers according to synthetic strategy of Scheme II from 4-hydroxy-1-(3-chlorophenyl)butan-1-ol and the nucleoside derivative. [M+H]+ calcd for C22H25ClFN2O9P: 562.11; found: 562.2. 1H NMR (400MHz, DMSO-d6 ) 11.56 (s, 1H), 7.76 (d, J = 8.4, 1H), 7.45-7.35 (m, 4H), 6.15 (m, 1H), 5.66 (t, J = 4.4, 1H), 5.38-5.22 (m, 2H), 3.25 (bs, 1H), 2.95-2.88 (m, 1H), 2.81 (s, 1.5H), 2.79 (s, 1.5H), 2.14 (s, 3H), 2.05-1.60 (m, 4H), and 1.33 (d, J = 23, 3H).
  • Example 7 (1S,3R,7S,8S,8aR)-3,7-dimethyl-8-(2-((2S,4R)-4-((2-oxido-4,6-di(pyridin-3-yl)-1,5,3,2-dioxazaphosphinin-2-yl)oxy)-6-oxotetrahydro-2H-pyran-2-yl)ethyl)-1,2,3,7,8,8a-hexahydronaphthalen-1-yl 2,2-dimethylbutanoate (Compound 107)
  • Figure imgb0048
  • Compound 107 can be prepared according to synthetic strategy of Scheme II from nicotinic acid, pyridine-3-aldehyde, and simvastatin. MH+ = 691.30(Calc.).
  • Example 8 (2R,3R,4R,5R)-2-(((4-(3-Chlorophenyl)-2-oxido-1,3,2-dioxaphosphepan-2-yl)oxy)methyl)-5-(2,4-dioxo-3,4-dihydropyrimidin-1(2H)-yl)-4-fluoro-4-methyltetrahydrofuran-3-yl acetate (Compound 108)
  • Figure imgb0049
  • Compound 108 was prepared as a mixture of two diastereomers according to synthetic strategy of Scheme II from 4-hydroxy-1-(3-chlorophenyl)butan-1-ol and the nucleoside derivative. [M+H]+ calcd for C22H25ClFN2O9P: 547.1; found: 547.2. 1H NMR (400MHz, DMSO-d6 ) 11.58 (s, 1H), 7.75-7.38 (m, 5H), 6.05-5.20 (m, 4H), 4.43-4.03 (m, 5H), 2.14 (s, 3H), 2.10-1.90 (m, 3H), 1.34 (d, J = 22, 1.5H), and 1.27 (d, J = 22, 1.5H).
  • Example 9 2-Hydroxy-2-oxido-4H-benzo[d][1,3,2]dioxaphosphinin-4-yl acetate (Compound 109)
  • Figure imgb0050
  • Compound 109 was prepared according the general procedure described in Scheme IV as a diisopropylethylamine salt from 2-hydroxybenzaldehyde using benzyl as protection group. [M-H]+ calcd for C9H9O6P: 243.01; found: 243.00.
  • Example 10 2,4-Dihydroxy-6-phenethyl-1,5,2,4-dioxadiphosphinane 2,4-dioxide (Compound 110)
  • Figure imgb0051
  • Compound 110 was prepared according to the general procedure of Scheme V as a triethylamine salt from 3-phenylpropanal and methylenebis(phosphonate). [M+H]+ calcd for C10H14O6P2: 293.03; found: 293.05. 1H NMR (300MHz, CD3OD) 7.28-7.06 (m, 5H), 5.67-5.57 (m, 1H), 3.17 (q, J = 7.4, 6H), 2.76 (t, J = 8.1, 2H), 2.38 (t, J = 28, 2H), 2.07-1.92 (m, 2H), and 1.29 (t, J = 7.4, 9H).
  • Example 11 2,4-Dihydroxy-6-phenethyl-1,3,5,2,4-trioxadiphosphinane 2,4-dioxide (Compound 111)
  • Figure imgb0052
  • Compound 111 was prepared according to the general procedure of Scheme V as a triethylamine salt from 3-phenylpropanal and pyrophosphate. [M+H]+ calcd for C9H12O7P2: 295.01; found: 295.15. 1H NMR (400MHz, CD3OD) 7.30-7.12 (m, 5H), 5.33 (t, J = 4.5, 1H), 3.19 (q, J = 7.4, 6H), 2.75 (t, J = 7.5, 2H), 2.08-1.95 (m, 2H), and 1.30 (t, J = 7.4, 9H).
  • Example 12 1-((Hydroxy(phosphonooxy)phosphoryl)oxy)-3-phenylpropyl acetate (Compound 112)
  • Figure imgb0053
  • Compound 112 was prepared according to the general procedure of Scheme V as a triethylamine salt from 1-chloro-3-phenylpropyl acetate and pyrophosphate. [M+H]+ calcd for C11H16O7P2: 355.03; found: 356.90. 1H NMR (400MHz, CD3OD) 7.28-7.12 (m, 5H), 6.60-6.52 (m, 1H), 3.17 (q, J = 7.4, 12H), 2.80-2.72 (m, 2H), 2.16-2.04 (m, 2H), 2.04 (s, 3H), 1.30 (t, J = 7.4, 18H).
  • Example 13 3-Phenylpropane-1,1-diyl bis(trihydrogen diphosphate) (Compound 113)
  • Figure imgb0054
  • Compound 113 was prepared according to the general procedure of Scheme V as a diisopropylethylamine salt from 1-chloro-3-phenylpropyl acetate and pyrophosphate. [M+H]+ calcd for C9H16O14P4: 472.95; found: 473.00. 1H NMR (400MHz, CD3OD) 7.28-7.12 (m, 5H), 5.75-5.65 (m, 1H), 3.20 (q, J = 7.4, 6H), 2.77 (t, J = 7.5, 2H), 2.06-1.95 (m, 2H), 1.30 (t, J = 7.4, 9H).
  • Example 14 1-((Di-tert-butoxyphosphoryl)oxy)-3-phenylpropyl acetate) (Compound 114)
  • Figure imgb0055
  • Compound 114 was prepared according to the general procedure of Scheme V from 1-chloro-3-phenylpropyl acetate and di-tert-butyl tetrabutylamonium phosphate. [2M+H]+ calcd for C38H62O12P2: 773.37; found: 773.0.
  • Example 15 Tetraphenyl(3-phenylpropane-1,1-diyl)bis(phosphate) (Compound 115)
  • Figure imgb0056
  • Compound 115 was prepared according to the general procedure of Scheme V from 1,1-diiodo-3-phenylpropane and diphenyl silver phosphate. [M+H]+ calcd for C33H30O8P2: 617.14; found: 616.90. 1H NMR (300MHz, CDCl3) 7.40-7.03 (m, 25H), 6.48-6.40 (m, 1H), 2.66-2.60 (m, 2H), and 2.24-2.15 (m, 2H).
  • Example 16 Tetraethyl (3-phenylpropane-1,1-diyl) bis(phosphate) (Compound 116)
  • Figure imgb0057
  • Compound 116 was prepared according to the general procedure of Scheme V from 1,1-diiodo-3-phenylpropane and diethyl silver phosphate. [M+H]+ calcd for C17H30O8P2: 424.14; found: 424.3. 1H NMR (400MHz, CD3OD) 7.71 (dd, J = 6.0 and 3.6, 1H), 7.61 (t, J = 3.6, 1H), 7.30-7.21 (m, 3H), 5.95-5.89 (m, 1H), 4.22-4.13 (m, 8H), 2.78 (t, J = 7.8, 2H), 1.74-1.67 (m, 2H), and 1.34-1.25 (m, 12H).
  • Example 17 ((4,5-cis)-2-((Z)-Heptadec-8-en-1-yl)-1,3-dioxolane-4,5-diyl)dimethanol (Compound 117)
  • Figure imgb0058
  • Compound 117 was prepared according to the general procedure of Scheme VI from (Z)-9-octadecenal and meso-erythritol. 1H NMR (400MHz, CDCl3) 5.37 (bs, 2H), 4.55 (t, J = 5.2, 1H), 4.18 (dd, J = 10.8 and 5.2, 1H), 3.95-3.76 (m, 3H), 3.53-3.38 (m, 2H), 2.08-2.00 (m, 4H), 1.68-1.60 (m, 4H), 1.40-1.25 (m, 20H), and 0.91 (t, J = 6.4, 3H).
  • Example 18 (Z)-(2-(Heptadec-8-en-1-yl)-1,3-dioxolan-4-yl)methanol (Compound 118)
  • Figure imgb0059
  • Compound 118 was prepared as a mixture of stereoisomers according to the general procedure of Scheme VI from (Z)-9-octadecenal and glycerol. 1H NMR (400MHz, CDCl3) 3.37 (bs, 2H), 4.18-4.08 (m, 2H), 3.95-3.87 (m, 1H), 3.80-3.55 (m, 4H), 2.10-1.99 (m, 4H), 1.68-1.57 (m, 3H), 1.45-1.28 (m, 21H), and 0.91 (t, J = 6.8, 3H).
  • Example 19 (Z)-(2-(Heptadec-8-en-1-yl)-1,3-dioxolan-4-yl)methyl dimethyl phosphate (Compound 119)
  • Figure imgb0060
  • Compound 119 was prepared as a mixture of stereoisomers from Compound 118 and dimethyl chlorophosphate. 1H NMR (400MHz, CDCl3) 5.34-5.27 (m, 2H), 4.92 (t, J = 4.2, 0.5H), 4.82 (t, J = 4.2, 0.5H),4.28-3.81 (m, 5H), 3.73 (s, 3H), 3.71 (s, 3H), 1.98-1.90 (m, 3H), 1.62-1.53 (m, 3H), 1.28-1.17 (m, 22H), and 0.81 (t, J = 6.0, 3H).
  • Example 20 2-(Pyridin-3-yl)-1,3-dioxan-5-yl oleate (Compound 120) and (2-(pyridin-3-yl)-1,3-dioxolan-4-yl)methyl oleate (Compound 121)
  • Figure imgb0061
    Figure imgb0062
  • Compounds 120 and 121 were prepared as a mixture of stereoisomers from oleic acid, nicotinaldehyde, and glycerol. Briefly, nicotinaldehyde was condensed with glycerol in the presence of an acid catalyst to afford a mixture of 5-member-ring and 6-member-ring acetals that were treated with oleic chloride to form the corresponding ester Compounds 120 and 121. [M+H]+ calcd for C27H43NO4: 446.32; found: 446.4. 1H NMR (400MHz, CDCl3) 8.66-8.54 (m, 2H), 7.94 (t, J = 10, 1H), 7.46 (bs, 1H), 5.99 (s, 0.5H), 5.87 (s, 0.5H), 5.33 (bs, 2H), 4.55-3.81 (m, 5H), 2.40-2.25 (m, 2H), 2.02 (bs, 4H), 1.70-1.55 (m, 2H), 1.28-1.17 (m, 20H), and 0.88 (t, J = 6.0, 3H).
  • Example 21 N-(5,5-Dimethyl-2-oxido-1,3,2-dioxaphosphinan-2-yl)oleamide (Compound 122)
  • Figure imgb0063
  • Compound 122 was prepared from oleic acid, 2,2-dimethylpropane-1,3-diol, and phosphorus oxychloride. Briefly, 2,2-dimethylpropane-1,3-diol was treated with phosphorus oxychloride to form the corresponding cyclic chlorophosphate that was then reacted with oleic amide to provide Compound 122. [M+H]+ calcd for C23H44NO4P: 430.30; found: 430.6. 1H NMR (400MHz, CD3OD) 5.35 (bs, 2H), 4.40 (d, J = 10, 2H), 4.01 (dd, J = 20 and 10, 2H), 2.38-2.02 (m, 4H), 1.65-1.56 (m, 2H), 1.38-1.25 (m, 22H), and 0.92 (t, J = 7.2, 3H).
  • Example 22 N-(5,5-Dimethyl-2-oxido-4-phenyl-1,3,2-dioxaphosphinan-2-yl)oleamide (Compound 123)
  • Figure imgb0064
  • Compound 123 was prepared as a mixture of stereoisomers from oleic acid, 2,2-dimethyl-1-phenylpropane-1,3-diol, and phosphorus oxychloride in a reaction sequence same as that of Compound 122. [M+H]+ calcd for C29H48NO4P: 506.33; found: 506.4. 1H NMR (400MHz, CDCl3) 8.52 (d, J = 10.4, 1H), 7.35-7.30 (m, 5H), 5.82 (s, 1H), 5.28 (bs, 2H), 4.84 (d, J = 10, 1H), 3.92 (dd, J = 23 and 10, 1H), 2.32-2.26 (m, 2H), 1.96-1.89 (m, 4H), 1.60-1.53 (m, 2H), 1.30-1.15 (m, 20H), 1.04 (s, 3H), 0.82 (t, J = 6.8, 3H), and 0.76 (s, 3H).
  • Example 23 1-((5,5-Dimethyl-2-oxido-4-phenyl-1,3,2-dioxaphosphinan-2-yl)oxy)ethyl oleate (Compound 124)
  • Figure imgb0065
  • Compound 124 was prepared as a mixture of stereoisomers from oleic acid, 2,2-dimethyl-1-phenylpropane-1,3-diol, and phosphorus oxychloride. Briefly, oleic acid was converted to the acid chloride with treatment of oxalyl chloride in dichloromethane and then the oleic chloride was treated with paraldehyde and zinc chloride in acetonitrile at 60-65 °C to give 1-chloroethyl oleate. The oleate was then reacted with 2-chloro-5,5-dimethyl-4-phenyl-1,3,2-dioxaphosphinane 2-oxide prepared from 2,2-dimethyl-1-phenylpropane-1,3-diol, and phosphorus oxychloride to afford Compound 124 as the final product after chromatography purification. [M+Na]+ calcd for C31H51O6P: 573.33; found: 573.4. 1H NMR (400MHz, CDCl3) 7.32-7.17 (m, 5H), 6.67-6.61 (m, 1H), 5.37 (s, 1H), 5.28 (bs, 2H), 4.39 (d, J = 10.8, 1H), 3.89 (ddd, J = 24, 11.8, and 3.6, 1H), 2.32-2.26 (m, 2H), 1.96-1.89 (m, 3H), 1.60-1.50 (m, 5H), 1.26-1.12 (m, 21H), 0.95 (d, J = 8.8, 3H), 0.81 (t, J = 6.8, 3H), and 0.73 (s, 3H).
  • Biological examples
  • Examples of use of the method include the following. It will be understood that the following are examples and that the embodiments are not limited to these examples.
  • Example A: In Vitro Activation of Prodrug Analogues by Rat Liver Microsomes. Quantification by LC-MS/MS
  • Prodrug compounds are tested for activation to the active drug in reactions catalyzed by the microsomal fraction of rat liver.
  • Methods:
  • Prodrug compounds are tested for activation by liver microsomes isolated from rats induced with dexamethasone to enhance CYP3A4 activity. Reactions are conducted in 0.1 M KH2PO4, pH 7.4, in the presence of 2 mM NADPH and liver microsomes (1 mg/mL). Reaction mixtures are incubated for 5 min. in an Eppendorf Thermomixer 5436 (37 °C, speed 6). Reactions are terminated by the addition of 1.5 volumes of methanol. The resulting extracts are clarified by centrifugation at 14,000 rpm in an Eppendorf microfuge (20 min.). The supernatants (200 µL) are evaporated under vacuum and heat to dryness. The dried residue is reconstituted with 200 µL of water and the mixture is centrifuged for 10 min at 14,000 rpm. A mixture of 35 µL aliquot of supernatant and 35 µL of mobile phase A is analyzed by LC-MS/MS. The active compound is detected by using MS/MS mode and quantified based on comparison to a standard active compound.
  • Example B: Active compound Accumulation in Hepatocytes Following Incubation with Prodrug compounds
  • Prodrug compounds are evaluated for their ability to generate active compounds in freshly isolated rat hepatocytes.
  • Methods:
  • Hepatocytes are prepared from fed Sprague-Dawley rats (250-300g) according to the procedure of Berry and Friend (Berry, M.N. Friend, D.S., J. Cell Biol. 43:506-520 (1969)) as modified by Groen (Groen, A.K. et al., Eur. J. Biochem 122:87-93 (1982)). Hepatocytes (20 mg/mL wet weight, >85% trypan blue viability) are incubated at 37 °C in 2 mL of Krebs-bicarbonate buffer containing 20 mM glucose, and 1 mg/mL BSA for 2 h in the presence of 1-250 µM a prodrug compound (from 25 mM stock solutions in DMSO). Following the incubation, 1600 µL aliquot of the cell suspension is centrifuged and 300 µL of acetonitrile is added to the pellet, vortexed and sonicated until the pellet broke down. Then 200 µL of water is added to make a 60% acetonitrile solution. After 10 min centrifugation at 14,000 rpm, the resulting supernatant is transferred to a new vial and evaporated to near dryness in a Savant SpeedVac Plus at room temperature. The dried residue is reconstituted with 200 µL of water and the mixture was centrifuged for 10 min at 14,000 rpm. A mixture of 35 µL aliquot of supernatant and 35 µL of mobile phase A (20 mM N-N-dimethylhexylamine and 10 mM propionic acid in 20% methanol) is analyzed by LC-MS/MS. The active compound is detected by using MS/MS mode (M-/78.8) and quantified based on comparison to a standard active compound.
  • Example C: Tissue Distribution Following Oral Administration of active compounds and their Prodrugs
  • The liver specificity of prodrugs is compared relative to their parent active compound in liver and other organs that could be targets of toxicity.
  • Methods:
  • Nucleoside analogues and their prodrugs are administered at 5-20 mg/kg to fasted rats by oral gavage. Plasma and portal vein concentrations of the active and prodrug are determined by HPLC-UV, and the liver, skeletal muscle, cardiac, kidney, small intestine, and other organ concentrations are measured by LC-MS using the standard chromatography method. The results demonstrate the liver targeting of the prodrug compounds and provide evidence for improved safety of the prodrugs over that of the actives. This can occur solely by the liver targeting provided by the prodrug.
  • All numbers expressing quantities of ingredients, reaction conditions, and so forth used in the specification are to be understood as being modified in all instances by the term "about." Accordingly, unless indicated to the contrary, the numerical parameters set forth herein are approximations that may vary depending upon the desired properties sought to be obtained. At the very least, and not as an attempt to limit the application of the doctrine of equivalents to the scope of any claims in any application claiming priority to the present application, each numerical parameter should be construed in light of the number of significant digits and ordinary rounding approaches.
  • Language of degree used herein, such as the terms "approximately," "about," "generally," and "substantially" as used herein represent a value, amount, or characteristic close to the stated value, amount, or characteristic that still performs a desired function or achieves a desired result. For example, the terms "approximately", "about", "generally," and "substantially" may refer to an amount that is within less than 10% of, within less than 5% of, within less than 1% of, within less than 0.1% of, and within less than 0.01% of the stated amount. As another example, in certain embodiments, the terms "generally parallel" and "substantially parallel" refer to a value, amount, or characteristic that departs from exactly parallel by less than or equal to 15°, 10°, 5°, 3°, 1°, 0.1°, or otherwise. Similarly, in certain embodiments, the terms "generally perpendicular" and "substantially perpendicular" refer to a value, amount, or characteristic that departs from exactly perpendicular by less than or equal to 15°, 10°, 5°, 3°, 1°, 0.1°, or otherwise.
  • The above description discloses several methods and materials. This invention is susceptible to modifications in the methods and materials, as well as alterations in the fabrication methods and equipment. Such modifications will become apparent to those skilled in the art from a consideration of this disclosure or practice of the invention disclosed herein. Consequently, it is not intended that this invention be limited to the specific embodiments disclosed herein, but that it cover all modifications and alternatives coming within the true scope and spirit of the invention.
  • All references cited herein, including but not limited to published and unpublished applications, patents, and literature references, are incorporated herein by reference in their entirety and are hereby made a part of this specification. To the extent publications and patents or patent applications incorporated by reference contradict the disclosure contained in the specification, the specification is intended to supersede and/or take precedence over any such contradictory material.
  • Aspects and features of the present disclosure are set out in the following numbered clauses which contain the subject matter of the claims of the parent application as filed.
    1. 1. A compound of Formula I:
      Figure imgb0066
      wherein:
      • R1 and R2 are each independently selected from the group consisting of H, M, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 acyloxy, an optionally substituted aryl, and an optionally substituted heteroaryl;
      • R3 is selected from the group consisting of H and an optionally substituted C1-C6 alkyl; or R3 optionally forms an optionally substituted ring with R2; or R3 together with R2 form a methylene or its derivative; or R3 together with R2 form an oxo (=O) or its derivative; or R3 optionally forms a bond with Z or Y' when Z or Y' is N;
      • M is a biological agent, or part of a biological agent or a prodrug of a biological agent;
      • X is selected from the group consisting of Cl, OR4, NR4R5, an optionally substituted C1-C6 alkyl, and M;
      • Y and Y' are each independently O or NR4; or Y' is CH2 or null;
      • Z is selected from the group consisting of O, NR5, CR8R6, C=O, C=NR7, and null; or Z is a 2-5 atom spacer selected from an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 heteroalkyl, an optionally substituted aryl, and an optionally substituted heteroaryl;
      • R4 is selected from the group of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
      • R5 is selected from the group consisting of H, M, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, and an optionally substituted C1-C6 acyl;
      • R6 is selected from the group consisting of F, Cl, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl; or R6 is H provided that R1 or R2 or R8 is connected to a cyclic core carbon atom through an oxygen-carbon (O-C) bond;
      • R7 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
      • R8 is selected from the group consisting of F, Cl, M, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl; or R8 is H provided that R1 or R2 is connected to a cyclic core carbon atom through an oxygen-carbon (O-C) bond;
      • provided that at least one of R1, R2, R5, R8, and X is M;
      • or a stereoisomer or a pharmaceutically acceptable salt thereof.
    2. 2. A compound of Formula Ia:
      Figure imgb0067
      wherein:
      • R1 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, an optionally substituted aryl, and an optionally substituted heteroaryl;
      • R2 and R3 are each independently selected from the group consisting of H and an optionally substituted C1-C6 alkyl; or R2 and R3 optionally form an optionally substituted ring; or R2 and R3 optionally form a methylene or its derivative; or R2 and R3 optionally form an oxo or its derivative;
      • M is a biological agent, part of a biological agent or a prodrug of a biological agent;
      • Y and Y' are each independently O or NR4; or Y' is CH2;
      • Z is selected from the group consisting of O, NR5, CR8R6, C=O, C=NR7, and null; or Z is a 2-5 atom spacer selected from an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 heteroalkyl, an optionally substituted aryl, and an optionally substituted heteroaryl;
      • R4 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
      • R5 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, and an optionally substituted C1-C6 acyl;
      • R6 is selected from the group consisting of F, Cl, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl; or R6 is H provided that R1 or R2 or R8 is connected with the cyclic core carbon atom through an oxygen-carbon (O-C) bond;
      • R7 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
      • R8 is selected from the group consisting of F, Cl, M, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl; or R8 is H provided that R1 or R2 is connected to a cyclic core carbon atom through an oxygen-carbon (O-C) bond;
      • or a stereoisomer or a pharmaceutically acceptable salt thereof.
    3. 3. A compound of Formula Ib:
      Figure imgb0068
      wherein:
      • R2 and R3 are independently selected from the group consisting of H and an optionally substituted C1-C6 alkyl; or R2 and R3 optionally form an optionally substituted ring; or R2 and R3 optionally form a methylene or its derivative; or R2 and R3 optionally form an oxo (=O) or its derivative;
      • M is a biological agent, part of a biological agent or a prodrug of a biological agent;
      • X is selected from the group consisting of Cl, an optionally substituted C1-C6 alkyl, NR4R5, and OR4;
      • Y and Y' are each independently O or NR4; or Y' is CH2 or null;
      • Z is selected from the group consisting of O, NR5, C(R6)2, C=O, C=NR7, and null; or Z is a 2-5 atom spacer selected from an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 heteroalkyl, an optionally substituted aryl, and an optionally substituted heteroaryl;
      • R4 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
      • R5 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, and an optionally substituted C1-C6 acyl;
      • R6 is selected from the group consisting of F, Cl, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl; or R6 is H provided that M or R2 is connected with the cyclic core carbon atom through an oxygen-carbon (O-C) bond;
      • R7 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
      • or a stereoisomer or a pharmaceutically acceptable salt thereof.
    4. 4. A compound of Formula Ic:
      Figure imgb0069
      wherein:
      • R1 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, an optionally substituted aryl, and an optionally substituted heteroaryl;
      • R3 is selected from the group consisting of H and an optionally substituted C1-C6 alkyl; or R3 together with M form a methnylene derivative; or R3 optionally forms a bond with Z or Y' when Z or Y' is N;
      • M is a biological agent, part of a biological agent or a prodrug of a biological agent;
      • X is selected from the group consisting of Cl, OR4, NR4R5, and an optionally substituted C1-C6 alkyl;
      • Y and Y' are each independently O or NR4;
      • Z is selected from the group consisting of O, NR5, C(R6)2, C=O, C=NR7, and null; or Z is a 2-5 atom spacer selected from an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 heteroalkyl, an optionally substituted aryl, and an optionally substituted heteroaryl;
      • R4 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
      • R5 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, and an optionally substituted C1-C6 acyl;
      • R6 is selected from the group consisting of F, Cl, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl; or R6 is H provided that R1 or M is connected with the cyclic core carbon atom through an oxygen-carbon (O-C) bond;
      • R7 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
      • or a stereoisomer or a pharmaceutically acceptable salt thereof.
    5. 5. A compound of Formula Id or Ie:
      Figure imgb0070
      wherein:
      • R2 and R3 are independently selected from the group consisting of H and an optionally substituted C1-C6 alkyl; or R3 together with M or R2 form a methylene derivative; or R3 together with M or R2 form an optionally substituted ring; or R3 together with R2 form an oxo (=O) or its derivative; or R3 optionally forms a bond with Z or Y' when Z or Y' is N;
      • M is a biological agent, part of a biological agent or a prodrug of a biological agent;
      • X is selected from the group consisting of Cl, OR4, and an optionally substituted C2-C6 alkyl;
      • Y and Y' are each independently O or N; or Y' is CH2;
      • Z is selected from the group consisting of O and NR5;
      • R4 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
      • R5 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, and an optionally substituted C1-C6 acyl;
      • or a stereoisomer or a pharmaceutically acceptable salt thereof.
    6. 6. A compound of Formula IIa or IIb:
      Figure imgb0071
      Figure imgb0072
      wherein:
      • R21 is selected from the group consisting of H, M, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 acyloxy, an optionally substituted aryl, and an optionally substituted heteroaryl;
      • M is a biological agent, or part of a biological agent or a prodrug of a biological agent;
      • Q is an optionally substituted aryl or an optionally substituted heteroaryl;
      • X2 is selected from the group consisting of Cl, OR24, N(R24)2 an optionally substituted C2-C6 alkyl, and M;
      • X'2 is selected from the group consisting of Cl, N(R24)2, and OR24;
      • Y2 and Y'2 are each independently O or NR24;
      • R24 is selected from the group of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
      • provided that at least one of R21 and X2 is M;
      • or a stereoisomer or a pharmaceutically acceptable salt thereof.
    7. 7. A compound of Formula III:
      Figure imgb0073
      wherein:
      • R31 is H; or R31 optionally forms a bond with M or X3 when X3 is N;
      • R32 and R33 are each independently selected from the group consisting of Cl, OH, NH2, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, and an optionally substituted C1-C6 alkylamino;
      • M is a biological agent or part of a biological agent or a prodrug of a biological agent;
      • X3 is O or NR34;
      • Y3 is selected from the group consisting of O, NR34, and an optionally substituted C1-C6 alkyl;
      • R34 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
      • or a stereoisomer or a pharmaceutically acceptable salt thereof.
    8. 8. A compound of Formula IV:
      Figure imgb0074
      wherein:
      • R41 is H; or R41 optionally forms a bond with M or X4 when X4 is N;
      • Z4 is selected from the group consisting of CR46R47, C(O), C(O)O, C(O)NR48, SO2, an optionally substituted aryl, and an optionally substituted heteroaryl;
      • R42, R43, R44, R45, R46, and R47 are each independently selected from the group consisting of H, OH, amino, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 acyloxy, an optionally substituted C1-C6 hereroalkyl, an optionally substituted phosphate, an optionally substituted phosphonate, an optionally substituted aryl, and an optionally substituted heterocycle; or R44 and R45 are independently or together optionally linked with R42, R43, R46, or R47 to form an optionally substituted ring; or R44 is optionally M; or R44 and R45 are together optionally to form an oxo (=O) or its derivative;
      • M is a biological agent or part of a biological agent or a prodrug of a biological agent;
      • X4 is selected from the group consisting of O, NR48, NC(O)R48, NS(O)2R49, and NP(O)(R50)2;
      • R48 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
      • R49 is selected from the group consisting of NH2, an optionally substituted C1-C6 alkyl, an optionally substituted aryl, and an optionally substituted heteroaryl;
      • R5 is selected from the group consisting of OH, NH2, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, and an optionally substituted C1-C6 alkylamino;
      • n is 0, 1, 2, or 3;
      • or a stereoisomer or a pharmaceutically acceptable salt thereof.
    9. 9. A compound of Formula Va, Vb, or Vc:
      Figure imgb0075
      wherein:
      • R51 and R52 are each independently selected from the group consisting of H, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 acyloxy, an optionally substituted C1-C6 acylamino, an optionally substituted phosphate, an optionally substituted phosphonate, an optionally substituted phosphoramidate, an optionally substituted C1-C6 aryl, and an optionally substituted heteroaryl; or R51 and R52 are together optionally to form an oxo (=O) or its derivative;
      • R53 and R54 are each independently selected from the group consisting of Cl, OH, NH2, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 alkylamino, an optionally substituted C1-C6 acylamino, an optionally substituted aryloxy, an optionally substituted phosphate, an optionally substituted phosphonate, and an optionally substituted heteroaryloxy; or R53 is optionally linked with R51, R54, or R56 to form an optionally substituted 5-, 6-, or 7-membered heterocycle;
      • R55 is selected from the group consisting of OH, NH2, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 alkylamino, an optionally substituted aryl, and an optionally substituted heteroaryl; or R55 is optionally linked to R51 or R56 to form an optionally substituted ring;
      • R56 is selected from the group consisting of H, a C1-C6 alkyl, and a C1-C6 heteroalkyl;
      • M is a biological agent or part of a biological agent or a prodrug of a biological agent;
      • X5 is O or NR56;
      • n is 0, 1, 2, or 3;
      • or a stereoisomer or a pharmaceutically acceptable salt thereof.
    10. 10. A compound of Formula VIa, VIb or VIc:
      Figure imgb0076
      wherein:
      • R61 and R62 are each independently selected from the group consisting of H, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 alkylamino, an optionally substituted C1-C6 acyloxy, -OCH2P(O)(R69)2, and an optionally substituted C1-C6 acylamino; or R61 and R62 together optionally form an oxo (=O) or its derivative;
      • R63, R64, and R65 are each independently selected from the group consisting of H, CO2R67, C(O)N(R67)2, P(O)(R69)2, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 acyloxy, and an optionally substituted C1-C6 hereroalkyl; or two of R63, R64, and R65 are optionally linked to form an optionally substituted ring; or R63 is optionally linked with R68 to form an optionally substituted ring; with the proviso that CR63R64R65 is not a straight chain C1-C4 alkyl when R61 and R62 form an oxo (=O);
      • R66 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 heteroalkyl, an optionally substituted aryl, and an optionally substituted heteroaryl;
      • R67 and R68 are each independently selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
      • R69 is selected from the group consisting of OH, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, and an optionally substituted C1-C6 alkylamino;
      • M is a biological agent or part of a biological agent or a prodrug of a biological agent;
      • X6 is O or NR68;
      • Y6 is selected from the group consisting of null, O, NR68, and C(R68)2;
      • or a stereoisomer or a pharmaceutically acceptable salt thereof.
    11. 11. A compound of Formula VII:
      Figure imgb0077
      wherein:
      • R71 is selected from the group consisting of H, OH, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 heteroalkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted phosphate, and an optionally substituted phosphonate;
      • X7 is O or S;
      • Y7 is Nor CR71;
      • M is a biological agent or part of a biological agent or a prodrug of a biological agent;
      • or a stereoisomer or a pharmaceutically acceptable salt thereof.
    12. 12. The compound of Clause 1, wherein the compound is selected from the group consisting of:
      Figure imgb0078
      Figure imgb0079
      Figure imgb0080
      Figure imgb0081
      and a stereoisomer or a pharmaceutically acceptable salt thereof.
    13. 13. The compound of any of Clauses 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or 11, wherein M is a nucleoside antiviral or anticancer agent.
    14. 14. The compound of any of Clauses 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or 11, wherein M is a lipid modulator.
    15. 15. The compound of any of Clauses 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or 11, wherein M is selected from the group consisting of HMG-CoA reductase inhibitor, a selective thyroid hormone receptor modulator, a peroxisome proliferator-activated receptor modulator, a fibrate, a nicotinic acid, a bile acid, and an omega-3 fatty acid.
    16. 16. The compound of any of Clauses 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or 11, wherein M is a glucose modulator.
    17. 17. The compound of any of Clauses 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or 11, wherein M is selected from the group consisting of a peroxisome proliferator-activated receptor modulator, a glucose biosynthesis inhibitor, and a dipeptidyl peptidase 4 inhibitor.
    18. 18. The compound of any of Clauses 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or 11, wherein M is a nuclear hormone receptor modulator.
    19. 19. A pharmaceutical composition comprising the compound of any one of Clauses 1-18 and a pharmaceutically acceptable excipient.
    20. 20. A method of treating a disease, disorder or condition comprising administering an effective amount of the compound of any one of Clauses 1-18 to a subject in need thereof.
    21. 21. The method of Clause 20, wherein the disease, disorder or condition is a disease, disorder or condition of the liver.
    22. 22. The method of Clause 20, wherein the disease, disorder or condition is a metabolic, cardiovascular or hormonal disease in which the liver is involved in the production and/or the homeostasis control of the biochemical end products of the disease, disorder or condition.
    23. 23. The method of Clause 20, wherein the disease, disorder or condition is selected from the group consisting of hepatitis, liver cancer, liver fibrosis, fatty liver, malaria, viral infection, parasitic infection, diabetes, hyperlipidemia, atherosclerosis, obesity, dyslipidemia, hyperglycemia and a hormonal condition.
    24. 24. A method of treating a liver disease comprising administering an effective amount of a compound of any one of Clauses 1-11 to a subject in need thereof,
      wherein M is a nucleoside antiviral or anticancer agent.
    25. 25. A method of treating dyslipidemia comprising administering to a subject in need thereof an effective amount of the compound of any one of Clauses 1-11,
      wherein M is a lipid modulator.
    26. 26. The method of Clause 24, wherein M is selected from the group consisting of HMG-CoA reductase inhibitor, a selective thyroid hormone receptor modulator, peroxisome proliferator-activated receptor modulator, a fibrate, nicotinic acid, a bile acid, and an omega-3 fatty acid.
    27. 27. A method of treating hyperglycemia comprising administering to a subject in need thereof, an effective amount of the compound of any one of Clauses 1-11,
      wherein M is a glucose modulator.
    28. 28. The method of Clause 27, wherein M is selected from the group consisting of a peroxisome proliferator-activated receptor modulator, a glucose biosynthesis inhibitor, and a dipeptidyl peptidase 4 inhibitor.
    29. 29. A method of treating a hormonal condition comprising administering to a subject in need thereof, an effective amount of a compound of any one of Clauses 1-11,
      wherein M is a nuclear hormone receptor modulator.
    30. 30. The method of any one of Clauses 20-29, further comprising administering an effective amount of at least one additional therapeutic agent to the subject in need thereof.
    31. 31. A method of delivering a diagnostic imaging agent to the liver of a subject in need thereof, comprising administering to the subject an effective amount of a compound of any one of Clauses 1-18.
    32. 32. The method of any one of Clauses 20-29, wherein the subject is a mammal.
    33. 33. The method of any one of Clauses 20-29, wherein the subject is human.
    34. 34. A method of inhibiting viral replication in a cell comprising contacting the cell with any one of the compounds of Clauses 1-18.
    35. 35. A method of intervening in a molecular pathway or modulating a target in a cell comprising contacting the cell with any one of the compounds of Clauses 1-18.
    36. 36. The method of Clauses 34 or 35, wherein the cell is in vivo.
    37. 37. The method of Clauses 34 or 35, wherein the cell is ex vivo.
    38. 38. The method of Clauses 34 or 35, wherein the cell is a hepatocyte.
    39. 39. The method of Clauses 34 or 35, wherein the cell is mammalian.
    40. 40. The method of Clauses 34 or 35, wherein the cell is human.

Claims (11)

  1. A compound of Formula IV:
    Figure imgb0082
    wherein:
    R41 is H; or R41 optionally forms a bond with M or X4 when X4 is N;
    Z4 is selected from the group consisting of CR46R47, C(O), C(O)O, C(O)NR48, SO2, an optionally substituted aryl, and an optionally substituted heteroaryl;
    R42, R43, R44, R45, R46, and R47 are each independently selected from the group consisting of H, OH, amino, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, an optionally substituted C1-C6 acyloxy, an optionally substituted C1-C6 hereroalkyl, an optionally substituted phosphate, an optionally substituted phosphonate, an optionally substituted aryl, and an optionally substituted heterocycle; or R44 and R45 are independently or together optionally linked with R42, R43, R46, or R47 to form an optionally substituted ring; or R44 is optionally M; or R44 and R45 are together optionally to form an oxo (=O) or its derivative;
    M is a biological agent or part of a biological agent or a prodrug of a biological agent;
    X4 is selected from the group consisting of O, NR48, NC(O)R48, NS(O)2R49, and NP(O)(R50)2;
    R48 is selected from the group consisting of H, an optionally substituted C1-C6 alkyl, and an optionally substituted C1-C6 heteroalkyl;
    R49 is selected from the group consisting of NH2, an optionally substituted C1-C6 alkyl, an optionally substituted aryl, and an optionally substituted heteroaryl;
    R50 is selected from the group consisting of OH, NH2, an optionally substituted C1-C6 alkyl, an optionally substituted C1-C6 alkyloxy, and an optionally substituted C1-C6 alkylamino;
    n is 0, 1, 2, or 3;
    or a stereoisomer or a pharmaceutically acceptable salt thereof.
  2. The compound of Claim 1, wherein M is:
    a) a nucleoside antiviral or nucleoside anticancer agent; or
    b) a lipid modulator; or
    c) selected from the group consisting of HMG-CoA reductase inhibitor, a selective thyroid hormone receptor modulator, a peroxisome proliferator-activated receptor modulator, a fibrate, a nicotinic acid, a bile acid, and an omega-3 fatty acid; or
    d) a glucose modulator; or
    e) selected from the group consisting of a peroxisome proliferator-activated receptor modulator, a glucose biosynthesis inhibitor, and a dipeptidyl peptidase 4 inhibitor; or
    f) a nuclear hormone receptor modulator.
  3. A pharmaceutical composition comprising the compound of Claim 1 or Claim 2 and a pharmaceutically acceptable excipient.
  4. A compound of Claim 1 or Claim 2 for use in medicine.
  5. The compound for use of Claim 4, wherein the compound is for use in treating a disease, disorder or condition, wherein the disease, disorder or condition is:
    a) a disease, disorder or condition of the liver; or
    b) a metabolic, cardiovascular or hormonal disease in which the liver is involved in the production and/or the homeostasis control of the biochemical end products of the disease, disorder or condition; or
    c) selected from the group consisting of hepatitis, liver cancer, liver fibrosis, fatty liver, malaria, viral infection, parasitic infection, diabetes, hyperlipidemia, atherosclerosis, obesity, dyslipidemia, hyperglycemia and a hormonal condition.
  6. A compound of Claim 1 or Claim 2 for use in:
    a) a method of treating a liver disease,
    wherein M is a nucleoside antiviral or anticancer agent, optionally wherein M is selected from the group consisting of HMG-CoA reductase inhibitor, a selective thyroid hormone receptor modulator, peroxisome proliferator-activated receptor modulator, a fibrate, nicotinic acid, a bile acid, and an omega-3 fatty acid; or
    b) a method of treating dyslipidemia,
    wherein M is a lipid modulator; or
    c) a method of treating hyperglycemia,
    wherein M is a glucose modulator, optionally wherein M is selected from the group consisting of a peroxisome proliferator-activated receptor modulator, a glucose biosynthesis inhibitor, and a dipeptidyl peptidase 4 inhibitor; or
    d) a method of treating a hormonal condition,
    wherein M is a nuclear hormone receptor modulator.
  7. The compound for use of Claim 5 or Claim 6, wherein the compound is administered in combination with at least one additional therapeutic agent.
  8. A compound of Claim 1 or Claim 2 for use in a method of delivering a diagnostic imaging agent to the liver of a subject in need thereof.
  9. The compound for use of Claim 5 or Claim 6, wherein the subject is a mammal, optionally wherein the subject is human.
  10. A compound of Claim 1 or Claim 2 for use in:
    a) a method of inhibiting viral replication in a cell; or
    b) a method of intervening in a molecular pathway or modulating a target in a cell; optionally wherein the cell is:
    a) a hepatocyte; or
    b) mammalian, optionally wherein the cell is human.
  11. An ex vivo method of:
    a) inhibiting viral replication in a cell comprising contacting the cell with any one of the compounds of Claim 1 or Claim 2.
    b) intervening in a molecular pathway or modulating a target in a cell comprising contacting the cell with any one of the compounds of Claim 1 or Claim 2; optionally wherein the cell is:
    a) a hepatocyte; or
    b) mammalian, optionally wherein the cell is human.
EP19207646.1A 2014-02-13 2015-02-11 Prodrug compounds and their uses Pending EP3623364A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201461939615P 2014-02-13 2014-02-13
US201461988101P 2014-05-02 2014-05-02
US201461988118P 2014-05-02 2014-05-02
EP15748902.2A EP3105238A4 (en) 2014-02-13 2015-02-11 Prodrug compounds and their uses
PCT/US2015/015496 WO2015123352A1 (en) 2014-02-13 2015-02-11 Prodrug compounds and their uses

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
EP15748902.2A Division EP3105238A4 (en) 2014-02-13 2015-02-11 Prodrug compounds and their uses

Publications (1)

Publication Number Publication Date
EP3623364A1 true EP3623364A1 (en) 2020-03-18

Family

ID=53800601

Family Applications (2)

Application Number Title Priority Date Filing Date
EP19207646.1A Pending EP3623364A1 (en) 2014-02-13 2015-02-11 Prodrug compounds and their uses
EP15748902.2A Withdrawn EP3105238A4 (en) 2014-02-13 2015-02-11 Prodrug compounds and their uses

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP15748902.2A Withdrawn EP3105238A4 (en) 2014-02-13 2015-02-11 Prodrug compounds and their uses

Country Status (7)

Country Link
US (2) US10449210B2 (en)
EP (2) EP3623364A1 (en)
JP (3) JP2017512183A (en)
CN (2) CN114404427A (en)
AU (3) AU2015217221A1 (en)
CA (1) CA2937548C (en)
WO (1) WO2015123352A1 (en)

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10449210B2 (en) 2014-02-13 2019-10-22 Ligand Pharmaceuticals Inc. Prodrug compounds and their uses
EP3164136A4 (en) * 2014-07-02 2018-04-04 Ligand Pharmaceuticals, Inc. Prodrug compounds and uses therof
ITUB20161079A1 (en) * 2016-02-25 2017-08-25 Hc Pharma Ag PROCEDURE FOR THE PREPARATION OF SOFOSBUVIR
CN109311925B (en) * 2016-05-12 2022-06-03 罗氏创新中心哥本哈根有限公司 Enhanced coupling of sterically defined oxaazaphospholane phosphoramidite monomers to nucleosides or oligonucleotides
CA3087932A1 (en) 2018-01-09 2019-07-18 Ligand Pharmaceuticals, Inc. Acetal compounds and therapeutic uses thereof
EP3788053B1 (en) * 2018-05-04 2024-07-10 Amgen Inc. Kras g12c inhibitors and methods of using the same
CN114846001B (en) * 2019-12-27 2024-01-23 南京明德新药研发有限公司 Benzo condensed ring compound

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001019829A2 (en) * 1999-09-17 2001-03-22 Basf Aktiengesellschaft Pyrazolopyrimidines as therapeutic agents
US6312662B1 (en) 1998-03-06 2001-11-06 Metabasis Therapeutics, Inc. Prodrugs phosphorus-containing compounds
US20060014740A1 (en) * 2003-11-18 2006-01-19 Miller Duane D Analogs exhibiting inhibition of cell proliferation, methods of making, and uses thereof
US7205404B1 (en) 1999-03-05 2007-04-17 Metabasis Therapeutics, Inc. Phosphorus-containing prodrugs
WO2008116156A2 (en) * 2007-03-21 2008-09-25 The Regents Of The University Of Michigan Methods and compositions for treating diseases and conditions associated with mitochondrial function
WO2009011850A2 (en) * 2007-07-16 2009-01-22 Abbott Laboratories Novel therapeutic compounds
WO2009073506A2 (en) 2007-11-29 2009-06-11 Metabasis Therapeutics Inc. Nucleoside prodrugs and uses thereof
US7666855B2 (en) 2004-02-13 2010-02-23 Metabasis Therapeutics, Inc. 2′-C-methyl nucleoside derivatives
WO2011084402A1 (en) * 2009-12-21 2011-07-14 Merck Sharp & Dohme Corp. Tyrosine kinase inhibitors
US8063025B2 (en) 2007-11-29 2011-11-22 Metabasis Therapeutics, Inc. Nucleoside prodrugs and uses thereof
WO2013074386A2 (en) * 2011-11-15 2013-05-23 Merck Sharp & Dohme Corp. Hcv ns3 protease inhibitors

Family Cites Families (281)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NL99649C (en) 1956-12-20
US3116282A (en) 1960-04-27 1963-12-31 Upjohn Co Pyrimidine nucleosides and process
BE633481A (en) 1962-06-12
CA777769A (en) 1963-03-18 1968-02-06 H. Roy Clarence Substituted methylene diphosphonic acid compounds and detergent compositions
US3328388A (en) 1964-09-02 1967-06-27 Merck & Co Inc Arabinofuranosyl pyrimidines and methods of preparing same
CH492738A (en) 1967-09-13 1970-06-30 Sandoz Ag Chlorethyl group-containing oxazaphosph- - orine derivatives
US3796700A (en) 1970-06-30 1974-03-12 Takedo Chem Ind Ltd Adenosine derivatives and the production thereof
DE2752125A1 (en) 1977-11-22 1979-05-23 Max Planck Gesellschaft NEW ORGANIC PHOSPHORUS RING COMPOUNDS, THEIR PRODUCTION AND USE
US4376165A (en) 1978-06-22 1983-03-08 Miles Laboratories, Inc. Method of preparing an enzyme-labeled ligand for use in specific binding assays and the labeled conjugate produced thereby
US4318982A (en) 1979-06-04 1982-03-09 Miles Laboratories, Inc. FMN-Labeled specific binding assay
US4340668A (en) 1979-06-04 1982-07-20 Miles Laboratories, Inc. Heme-labeled specific binding assay
US4255566A (en) 1980-02-19 1981-03-10 Miles Laboratories Flavin adenine dinucleotide derivatives and labeled conjugates prepared therefrom
US4447529A (en) 1981-07-06 1984-05-08 Miles Laboratories, Inc. Preparing homogeneous specific binding assay element to avoid premature reaction
DE3132221A1 (en) 1981-08-14 1983-05-19 Behringwerke Ag, 3550 Marburg NEW CYCLOPHOSPHAMIDE DERIVATIVES, METHOD FOR THE PRODUCTION THEREOF AND THEIR USE
DE3133077A1 (en) 1981-08-21 1983-03-10 Henkel KGaA, 4000 Düsseldorf NEW 1,3,2-OXAZAPHOSPHORINE COMPOUNDS CONTAINING NEW CYTOSTATICALLY EFFECTIVE EPOXY GROUPS, METHOD FOR THE PRODUCTION THEREOF AND THEIR USE IN PHARMACEUTICAL PREPARATIONS
IT1201087B (en) 1982-04-15 1989-01-27 Gentili Ist Spa PHARMACOLOGICALLY ACTIVE BIPPHOSPHONES, PROCEDURE FOR THEIR PREPARATION AND RELATED PHARMACEUTICAL COMPOSITIONS
US4440740A (en) 1982-04-26 1984-04-03 Merck & Co., Inc. α-Keto aldehydes as enhancing agents of gastro-intestinal drug absorption
US4464389A (en) 1982-10-20 1984-08-07 Sri International Esters of retinoic acid and pentaerythritol and monobenzal acetals thereof
CS233665B1 (en) 1983-01-06 1985-03-14 Antonin Holy Processing of isomere o-phosphonylmethylderivative of anantiomere racemic vicinal diene
US5047533A (en) 1983-05-24 1991-09-10 Sri International Acyclic purine phosphonate nucleotide analogs
DE3569737D1 (en) 1984-03-01 1989-06-01 Asta Pharma Ag Salts of oxazaphosphorine derivatives
US4579849A (en) 1984-04-06 1986-04-01 Merck & Co., Inc. N-alkylguanine acyclonucleosides as antiviral agents
FR2562543B1 (en) 1984-04-10 1987-09-25 Elf Aquitaine NOVEL CYCLIC PHOSPHONITES, THEIR PREPARATION AND APPLICATIONS
US4749694A (en) 1984-04-26 1988-06-07 Merck & Co., Inc. Novel lysine esters used as absorption
US4537772A (en) 1984-05-02 1985-08-27 Merck & Co., Inc. Enhancing absorption of drugs from gastrointestinal tract using acylcarnitines
US4898724A (en) 1984-06-04 1990-02-06 The Dow Chemical Company Organis amine phosphonic acid complexes for the treatment of calcific tumors
NL8403224A (en) 1984-10-24 1986-05-16 Oce Andeno Bv DIOXAPHOSPHORINANS, THEIR PREPARATION AND THE USE FOR SPLITTING OF OPTICALLY ACTIVE COMPOUNDS.
IT1196315B (en) 1984-10-29 1988-11-16 Gentili Ist Spa PROCEDURE FOR THE PREPARATION OF DIPHOSPHONIC ACIDS
IL77243A (en) 1984-12-21 1996-11-14 Procter & Gamble Pharmaceutical compositions containing geminal diphosphonic acid compounds and certain such novel compounds
US4724232A (en) 1985-03-16 1988-02-09 Burroughs Wellcome Co. Treatment of human viral infections
CS263951B1 (en) 1985-04-25 1989-05-12 Antonin Holy 9-(phosponylmethoxyalkyl)adenines and method of their preparation
CS263952B1 (en) 1985-04-25 1989-05-12 Holy Antonin Remedy with antiviral effect
US4973579A (en) 1985-06-28 1990-11-27 Merck & Co., Inc. Enhancment of absorption of drugs from gastrointestinal tract using choline ester salts
US4729989A (en) 1985-06-28 1988-03-08 Merck & Co., Inc. Enhancement of absorption of drugs from gastrointestinal tract using choline ester salts
US4822773A (en) 1985-06-28 1989-04-18 Merck & Co., Inc. Enhancement of absorption of drugs from gastrointestinal tract using choline ester salts
US4847298A (en) 1985-08-16 1989-07-11 Merck & Co., Inc. Acylcarnitines as absorption-enhancing agents for drug delivery through mucous membranes of the nasal, buccal, sublingual and vaginal compartments
US4963525A (en) 1985-08-16 1990-10-16 Merck & Co., Inc. Acylcarnitines as absorption-enhancing agents for drug delivery through mucous membranes of the nasal, buccal, sublingual and vaginal compartments
US4731360A (en) 1985-08-16 1988-03-15 Merck & Co., Inc. Acylcarnitines as absorption-enhancing agents for drug delivery through mucous membranes of the nasal, buccal, sublingual and vaginal compartments
US4692441A (en) 1985-08-16 1987-09-08 Merck & Co., Inc. Chorine esters as absorption-enhancing agents for drug delivery through mucous membranes of the nasal, buccal, sublingual and vaginal cavities
US4963556A (en) 1985-08-16 1990-10-16 Merck & Co., Inc. Choline esters as absorption-enhancing agents for drug delivery through mucous membranes of the nasal, buccal, sublingual and vaginal cavities
US5258538A (en) 1985-08-26 1993-11-02 Applied Biosystems, Inc. 2,3-disubstituted-1,3,2-oxazaphosphacycloalkanes as nucleic acid linking agents
US4879277A (en) 1985-08-26 1989-11-07 The United States Of America As Represented By The Department Of Health And Human Services Antiviral compositions and methods
US4757141A (en) 1985-08-26 1988-07-12 Applied Biosystems, Incorporated Amino-derivatized phosphite and phosphate linking agents, phosphoramidite precursors, and useful conjugates thereof
JPS62501712A (en) 1985-08-26 1987-07-09 アメリカ合衆国 Anti-HTLV-3/LAV agent containing 2',3'-dideoxyinosine, 2',3'-dideoxyguanosine or 2',3'-dideoxyadenosine
US5246937A (en) 1985-09-18 1993-09-21 Beecham Group P.L.C. Purine derivatives
DE3600289A1 (en) 1986-01-08 1987-07-09 Bayer Ag CYCLIC MALONYLPHOSPHONIC ACID DIAMOND, METHOD FOR THE PRODUCTION THEREOF AND THEIR USE AS A PEST CONTROL
JPH0615553B2 (en) 1986-02-20 1994-03-02 日産化学工業株式会社 Dihydropyridine-5-phosphonamidic acids
US4783443A (en) 1986-03-03 1988-11-08 The University Of Chicago Amino acyl cephalosporin derivatives
US4968788A (en) 1986-04-04 1990-11-06 Board Of Regents, The University Of Texas System Biologically reversible phosphate and phosphonate protective gruops
US4839466A (en) 1986-04-11 1989-06-13 The Rockefeller University Insulin activity messengers
JPS62249996A (en) 1986-04-23 1987-10-30 Asahi Chem Ind Co Ltd Novel benzoxazaphosphorine derivative and insecticidal composition containing said derivative as active ingredient
US5091552A (en) 1986-06-30 1992-02-25 Board Of Regents, The University Of Texas System Novel antitumor aldophosphamide analogs
ATE66609T1 (en) 1986-07-11 1991-09-15 Asta Pharma Ag OXAZAPHOSPHORINE SOLUTIONS WITH IMPROVED STABILITY AND PROCESSES FOR THEIR PREPARATION.
CS264222B1 (en) 1986-07-18 1989-06-13 Holy Antonin N-phosphonylmethoxyalkylderivatives of bases of pytimidine and purine and method of use them
DE3626058A1 (en) 1986-08-01 1988-02-11 Boehringer Mannheim Gmbh NEW DIPHOSPHONIC ACID DERIVATIVES, METHOD FOR THE PRODUCTION THEREOF AND MEDICINAL PRODUCTS CONTAINING THESE COMPOUNDS
JPS6360929A (en) 1986-08-29 1988-03-17 Yodogawa Seiyaku Kk Antitumor agent
ATE72816T1 (en) 1986-11-21 1992-03-15 Ciba Geigy Ag NEW SUBSTITUTE ALKANEDIPHOSPHONE ACIDS.
US5159067A (en) 1987-01-28 1992-10-27 University Of Georgia Research Foundation Inc. 5'-Diphosphohexose nucleoside pharmaceutical compositions
US5077280A (en) 1988-04-12 1991-12-31 Brown University Research Foundation Treatment of viral infections
EP0338372A3 (en) 1988-04-22 1991-10-09 American Cyanamid Company Solubilized pro-drugs
GB8815265D0 (en) 1988-06-27 1988-08-03 Wellcome Found Therapeutic nucleosides
ATE128623T1 (en) 1988-08-02 1995-10-15 Nissan Chemical Ind Ltd AGENT FOR IMPROVING DRUG EFFECTS FOR ANTITUMOR DRUGS.
US4882142A (en) 1988-12-19 1989-11-21 The Dow Chemical Company Bone marrow suppressing agents
US5658889A (en) 1989-01-24 1997-08-19 Gensia Pharmaceuticals, Inc. Method and compounds for aica riboside delivery and for lowering blood glucose
US5118672A (en) 1989-07-10 1992-06-02 University Of Georgia Research Foundation 5'-diphosphohexose nucleoside pharmaceutical compositions
JPH0377894A (en) 1989-08-18 1991-04-03 Toray Ind Inc Methylene diphosphonic acid compound
US5914331A (en) 1990-02-01 1999-06-22 Emory University Antiviral activity and resolution of 2-hydroxymethyl-5-(5-fluorocytosin-1-yl)-1,3-oxathiolane
US5204466A (en) 1990-02-01 1993-04-20 Emory University Method and compositions for the synthesis of bch-189 and related compounds
US5627165A (en) 1990-06-13 1997-05-06 Drug Innovation & Design, Inc. Phosphorous prodrugs and therapeutic delivery systems using same
CS387190A3 (en) 1990-08-06 1992-03-18 Ustav Organicke Chemie A Bioch (2r)-2-/di(2-propyl)phosphonylmethoxy/-3-p-toluenesulfonyloxy -1- trimethylacetoxypropane and process for preparing thereof
DE10399025I2 (en) 1990-09-14 2007-11-08 Acad Of Science Czech Republic Active substance precursors of phosphonates
HU206883B (en) 1990-12-18 1993-01-28 Richter Gedeon Vegyeszet Process for producing new 15-nitro-2-beta-, 3-beta-dihydro- and 15-nitro-2-beta, 3-beta-, 6,7-tetrahydro-tabersonin derivatives and pharmaceutical compositions containing them
US5168103A (en) 1991-01-22 1992-12-01 American Home Products Corporation [[2-(amino-3,4-dioxo-1-cyclobuten-1-yl) amino]alkyl]-acid derivatives
US5157027A (en) 1991-05-13 1992-10-20 E. R. Squibb & Sons, Inc. Bisphosphonate squalene synthetase inhibitors and method
JP2648897B2 (en) 1991-07-01 1997-09-03 塩野義製薬株式会社 Pyrimidine derivatives
FR2681784B1 (en) 1991-10-01 1995-06-09 Fabre Pierre Cosmetique DERMATOLOGICAL AND / OR COSMETOLOGICAL COMPOSITION CONTAINING RETINOUIDES AND USE OF NEW RETINOUIDES.
WO1993016388A1 (en) 1992-02-13 1993-08-19 The Uab Research Foundation Method of detecting and monitoring levels of 3'-amino-3'-deoxythymidine in body fluids and antibodies for same
GB2266525A (en) 1992-03-17 1993-11-03 Merck & Co Inc Substituted cephalosporin sulfone compositions useful in the treatment of leukemia
GB2266527A (en) 1992-03-17 1993-11-03 Merck & Co Inc Substituted azetidinones useful in the treatment of leukemia
GB9205917D0 (en) 1992-03-18 1992-04-29 Smithkline Beecham Plc Pharmaceuticals
US5480875A (en) 1992-06-23 1996-01-02 Yamanouchi Pharmaceutical Co., Ltd. Crystal of monohydrate of heterocyclic bis(phosphonic acid) derivative
US5532225A (en) 1992-07-31 1996-07-02 Sri International Acyclic purine phosphonate nucleotide analogs as antiviral agents, and related synthetic methods
US5358941A (en) 1992-12-02 1994-10-25 Merck & Co., Inc. Dry mix formulation for bisphosphonic acids with lactose
GB9301629D0 (en) 1993-01-27 1993-03-17 Scotia Holdings Plc Formulations containing unsaturated fatty acids
US5366965A (en) 1993-01-29 1994-11-22 Boehringer Mannheim Gmbh Regimen for treatment or prophylaxis of osteoporosis
RU2136693C1 (en) 1993-02-10 1999-09-10 Астра Фармасьютикалз Лимитед N-alkyl-substituted atp derivatives and method of preparation thereof
US5719303A (en) 1993-03-08 1998-02-17 Eisai Co., Ltd. Phosphonic acid derivatives
WO1994027590A1 (en) 1993-05-21 1994-12-08 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services New procedure to block the replication of reverse transcriptase dependent viruses by the use of inhibitors of deoxynucleotides synthesis
US5514798A (en) 1993-06-02 1996-05-07 Gilead Sciences, Inc. Method and cyclic carbonates for nucleotide analogues
DE69426904T2 (en) 1993-06-29 2001-10-11 Mitsubishi Chemical Corp., Tokio/Tokyo Phosphonate nucleotide ester derivatives
US5567689A (en) 1993-08-13 1996-10-22 The Uab Research Foundation Methods for increasing uridine levels with L-nucleosides
FR2709754B1 (en) 1993-09-10 1995-12-01 Centre Nat Rech Scient Compounds 2 'or 3'-deoxy- and 2', 3'-dideoxy-beta-L-pentofuranonucleosides, preparation process and therapeutic application, in particular anti-viral.
CA2637774C (en) 1993-09-10 2011-07-19 Emory University Nucleosides with anti-hepatitis b virus activity
US20020120130A1 (en) 1993-09-10 2002-08-29 Gilles Gosselin 2' or 3' -deoxy and 2', 3' -dideoxy-beta-L-pentofuranonucleo-side compounds, method of preparation and application in therapy, especially as anti- viral agents
AU691527B2 (en) 1993-09-17 1998-05-21 Gilead Sciences, Inc. Nucleotide analogs
US5437772A (en) 1993-11-01 1995-08-01 The Electrosynthesis Co., Inc. Portable lead detector
US5567592A (en) 1994-02-02 1996-10-22 Regents Of The University Of California Screening method for the identification of bioenhancers through the inhibition of P-glycoprotein transport in the gut of a mammal
US6143864A (en) 1994-06-28 2000-11-07 Merck & Co., Inc. Peptides
US5599686A (en) 1994-06-28 1997-02-04 Merck & Co., Inc. Peptides
US5866679A (en) 1994-06-28 1999-02-02 Merck & Co., Inc. Peptides
ATE199721T1 (en) 1994-07-04 2001-03-15 Takeda Chemical Industries Ltd PHOSPHONIC ACID COMPOUNDS IN PRODUCTION AND USE
US5810856A (en) 1995-03-09 1998-09-22 Tveras; Rimvydas Wiping element for an oral hygiene device, window wiper, or the like
US5665713A (en) 1995-04-12 1997-09-09 Procter & Gamble Company Pharmaceutical composition for inhibiting the growth of viruses and cancers
US5665386A (en) 1995-06-07 1997-09-09 Avmax, Inc. Use of essential oils to increase bioavailability of oral pharmaceutical compounds
CA2219132C (en) 1995-06-07 2007-03-06 Emory University Nucleosides with anti-hepatitis b virus activity
US5716928A (en) 1995-06-07 1998-02-10 Avmax, Inc. Use of essential oils to increase bioavailability of oral pharmaceutical compounds
US5627164A (en) 1995-06-13 1997-05-06 Exxon Research And Engineering Company Pyrazolyl borates complexes-(LAW294)
ES2210377T3 (en) 1995-07-17 2004-07-01 Cephalon, Inc. CISTEINE AND SERINA PROTEASA INHIBITORS CONTAINING PHOSPHORUS.
US5750493A (en) 1995-08-30 1998-05-12 Raymond F. Schinazi Method to improve the biological and antiviral activity of protease inhibitors
WO1997022614A1 (en) 1995-12-19 1997-06-26 Darwin Discovery Limited Ifosfamide, analogues thereof and their preparation
CA2195262C (en) 1996-01-18 2005-08-09 Masaru Ubasawa Phosphonate nucleotide compounds
US5767097A (en) 1996-01-23 1998-06-16 Icn Pharmaceuticals, Inc. Specific modulation of Th1/Th2 cytokine expression by ribavirin in activated T-lymphocytes
JPH09241284A (en) 1996-03-08 1997-09-16 Koji Yamashita New oxyazaphosphorine derivative
US6128582A (en) 1996-04-30 2000-10-03 Vertex Pharmaceuticals Incorporated Molecules comprising an IMPDH-like binding pocket and encoded data storage medium capable of graphically displaying them
US5962440A (en) 1996-05-09 1999-10-05 Bristol-Myers Squibb Company Cyclic phosphonate ester inhibitors of microsomal triglyceride transfer protein and method
RU2111970C1 (en) 1996-06-25 1998-05-27 Иван Игоревич Федоров 3'-oximino-2',3'-dideoxynucleosides
GB9618634D0 (en) 1996-09-06 1996-10-16 Univ Southampton Anti cancer drugs
US6177404B1 (en) 1996-10-15 2001-01-23 Merck & Co., Inc. Conjugates useful in the treatment of benign prostatic hyperplasia
SI20076A (en) 1996-10-16 2000-04-30 Icn Pharmaceuticals, Inc. Monocyclic l-nucleosides, analogs and use thereof
CA2269807C (en) 1996-10-24 2007-04-10 Novartis Ag Substituted aminoalkanephosphonic acids
US5948750A (en) 1996-10-30 1999-09-07 Merck & Co., Inc. Conjugates useful in the treatment of prostate cancer
GB9623908D0 (en) 1996-11-18 1997-01-08 Hoffmann La Roche Amino acid derivatives
CZ172799A3 (en) 1996-11-19 1999-10-13 Icn Pharmaceuticals, Inc. Polyvalent salts of pyridostigmine and related compounds
CA2278031A1 (en) 1996-12-27 1998-07-09 Robert Tam G-rich oligo aptamers and methods of modulating an immune response
SE9700792L (en) 1997-03-06 1998-02-23 Carlo Castro Back lubricant for lubricating skin cream on a human body
US6054587A (en) 1997-03-07 2000-04-25 Metabasis Therapeutics, Inc. Indole and azaindole inhibitors of fructose-1,6-bisphosphatase
ATE253073T1 (en) 1997-03-07 2003-11-15 Metabasis Therapeutics Inc NEW BENZIMIDAZOLE INHIBITORS OF FRUCTOSE-1,6-BISPHOSPHATASE
WO1998039344A1 (en) 1997-03-07 1998-09-11 Metabasis Therapeutics, Inc. Novel purine inhibitors of fructose-1,6-bisphosphatase
GB9707659D0 (en) 1997-04-16 1997-06-04 Peptide Therapeutics Ltd Hepatitis C NS3 Protease inhibitors
JP2002501546A (en) 1997-06-30 2002-01-15 アイ・シー・エヌ・フアーマシユーテイカルズ・インコーポレイテツド Methods for making thiazofurins and other C-nucleosides
US6130504A (en) 1997-07-11 2000-10-10 Sharp Kabushiki Kaisha Plasma addressing display device and method for producing the same
ES2187991T3 (en) 1997-07-25 2003-06-16 Gilead Sciences Inc NUCLEOTIDE ANALOG COMPOSITIONS.
IL121413A (en) 1997-07-28 2000-10-31 Green Cloud Ltd Method for reducing the accumulation of precipitates and impurities on ultrasonic transducers
US6767991B1 (en) 1997-08-11 2004-07-27 Boehringer Ingelheim (Canada) Ltd. Hepatitis C inhibitor peptides
US6143715A (en) 1997-08-11 2000-11-07 Boehringer Ingelheim (Canada) Ltd. Hepatitis C inhibitor peptide analogues
US6180666B1 (en) 1997-09-05 2001-01-30 Anmax, Inc. Use of gallic acid esters to increase bioavailability of orally administered pharmaceutical compounds
US5962522A (en) 1997-09-05 1999-10-05 Avmax, Inc. Propyl gallate to increase bioavailability of orally administered pharmaceutical compounds
US6391305B1 (en) 1997-09-10 2002-05-21 Merck & Co., Inc. Conjugates useful in the treatment of prostate cancer
US20020115596A1 (en) 1997-10-27 2002-08-22 Merk & Co., Inc. Conjugates useful in the treatment of prostate cancer
US6423695B1 (en) 1998-01-13 2002-07-23 Ribapharm, Inc. Cytokine related treatments of disease
WO1999036074A1 (en) 1998-01-15 1999-07-22 Icn Pharmaceuticals, Inc. Use of 8-cl-camp in prevention of restenosis of arterial walls
DK1058686T3 (en) 1998-02-25 2007-03-05 Univ Emory 2'-fluoro nucleosides
JP4741725B2 (en) 1998-03-06 2011-08-10 メタベイシス・セラピューティクス・インコーポレーテッド New prodrugs for phosphorus-containing compounds
CA2289621A1 (en) 1998-03-16 1999-09-23 Ontogen Corporation Piperazines as inhibitors of fructose-1,6-bisphosphatase (fbpase)
GB9806815D0 (en) 1998-03-30 1998-05-27 Hoffmann La Roche Amino acid derivatives
ES2281170T3 (en) 1998-03-31 2007-09-16 Vertex Pharmaceuticals Incorporated SERINA PROTEASES INHIBITORS, PARTICULARLY PROTEASE NS3 OF THE HEPATITIS VIRUS C.
IL139420A0 (en) 1998-05-26 2001-11-25 Icn Pharmaceuticals Novel nucleosides having bicyclic sugar moiety
GB9812523D0 (en) 1998-06-10 1998-08-05 Angeletti P Ist Richerche Bio Peptide inhibitors of hepatitis c virus ns3 protease
EP1089741A1 (en) 1998-06-24 2001-04-11 Emory University Use of 3'-azido-2',3'-dideoxyuridine in combination with further anti-hiv drugs for the manufacture of a medicament for the treatment of hiv
IT1305313B1 (en) 1998-07-17 2001-05-04 Colla Paolo 3,4 - DIHYDRO- 6- BENZYL-4-OXOPYRIMIDINE REPLACED AND RELATED PROCESS OF PRODUCTION AND USE IN THE THERAPY OF HIV-1 INFECTIONS.
US6444652B1 (en) 1998-08-10 2002-09-03 Novirio Pharmaceuticals Limited β-L-2'-deoxy-nucleosides for the treatment of hepatitis B
PT2415776T (en) 1998-08-10 2016-07-07 Novartis Ag Beta-l-2'-deoxy-nucleosides for the treatment of hepatitis b
KR20070053264A (en) 1998-09-09 2007-05-23 메타베이시스 테라퓨틱스, 인크. Novel heteroaromatic inhibitors of fructose 1,6-bisphosphatase
US6045638A (en) 1998-10-09 2000-04-04 Atlantic Research Corporation Monopropellant and propellant compositions including mono and polyaminoguanidine dinitrate
JP2002528499A (en) 1998-10-29 2002-09-03 ブリストル−マイヤーズ スクイブ カンパニー Amino nucleus-derived compounds that are inhibitors of the IMPDH enzyme
US6407077B1 (en) 1998-11-05 2002-06-18 Emory University β-L nucleosides for the treatment of HIV infection
AU774720B2 (en) 1998-11-05 2004-07-08 Centre National De La Recherche Scientifique Nucleosides with anti-hepatitis B virus activity
US6756360B1 (en) 1998-12-24 2004-06-29 Metabasis Therapeutics, Inc. Combination of FBPase inhibitors and insulin sensitizers for the treatment of diabetes
NZ512219A (en) 1998-12-24 2004-12-24 Metabasis Therapeutics Inc A combination of FBPase inhibitors and insulin sensitizers for the treatment of diabetes
US20020187945A1 (en) 1999-01-29 2002-12-12 Robert Tam Modulation of immune response by ribavirin
CZ20013180A3 (en) 1999-03-05 2002-02-13 Metabasis Therapeutics, Inc. Novel phosphorus-containing prodrugs
US6608027B1 (en) 1999-04-06 2003-08-19 Boehringer Ingelheim (Canada) Ltd Macrocyclic peptides active against the hepatitis C virus
WO2001000622A1 (en) 1999-06-25 2001-01-04 Vertex Pharmaceuticals Incorporated Prodrugs of carbamate inhibitors of impdh
RU2002103501A (en) 1999-08-27 2003-09-10 Ай-Си-Эн Фармасьютикалз, Инк. (Us) Pyrrolo [2,3-d] pyrimidine nucleoside analogues
US6752981B1 (en) 1999-09-08 2004-06-22 Metabasis Therapeutics, Inc. Prodrugs for liver specific drug delivery
US6921763B2 (en) 1999-09-17 2005-07-26 Abbott Laboratories Pyrazolopyrimidines as therapeutic agents
US6518253B1 (en) 1999-11-19 2003-02-11 Robert Tam Treatment of viral infections using the L-isomer of ribavirin
BR0016058A (en) 1999-12-03 2003-07-15 Univ California San Diego Phosphonate Compounds
AU5076601A (en) 1999-12-13 2001-07-03 Icn Pharmaceuticals, Inc. Pulmonary delivery of ribavirin or levovirinTM for systemic and quasi-systemic treatment of disease
CA2384326A1 (en) 1999-12-23 2001-06-28 Johnson Lau Compositions and methods for l-nucleosides, l-nucleotides, and their analogs
US6770666B2 (en) 1999-12-27 2004-08-03 Japan Tobacco Inc. Fused-ring compounds and use thereof as drugs
AU3653301A (en) 2000-01-28 2001-08-07 Merck & Co., Inc. Treatment or prevention of prostate cancer with a cox-2 selective inhibiting drug
US6455508B1 (en) 2000-02-15 2002-09-24 Kanda S. Ramasamy Methods for treating diseases with tirazole and pyrrolo-pyrimidine ribofuranosyl nucleosides
US6495677B1 (en) 2000-02-15 2002-12-17 Kanda S. Ramasamy Nucleoside compounds
JP2003522790A (en) 2000-02-17 2003-07-29 メルク エンド カムパニー インコーポレーテッド Treatment or prevention of prostate cancer using a COX-2 selective inhibitor
US6727267B2 (en) 2000-04-05 2004-04-27 Tularik Inc. NS5B HVC polymerase inhibitors
KR20100003313A (en) 2000-04-13 2010-01-07 파마셋 인코포레이티드 3'- or 2'-hydroxymethyl substituted nucleoside derivatives for treatment of hepatitis virus infections
MY164523A (en) 2000-05-23 2017-12-29 Univ Degli Studi Cagliari Methods and compositions for treating hepatitis c virus
EP1736478B1 (en) 2000-05-26 2015-07-22 IDENIX Pharmaceuticals, Inc. Methods and compositions for treating flaviviruses and pestiviruses
AU2001266610A1 (en) 2000-05-26 2001-12-11 Versatile Optical Networks, Inc. Wavelength locking system
WO2001091737A2 (en) 2000-05-26 2001-12-06 Idenix (Cayman) Limited Methods for treating hepatitis delta virus infection with beta-l-2' deoxy-nucleosides
US6875751B2 (en) 2000-06-15 2005-04-05 Idenix Pharmaceuticals, Inc. 3′-prodrugs of 2′-deoxy-β-L-nucleosides
US6545007B2 (en) 2000-11-17 2003-04-08 Idenix (Cayman) Limited Methods for inhibiting the transmission of HIV using topically applied substituted 6-benzyl-4-oxopyrimidines
GB0015627D0 (en) 2000-06-26 2000-08-16 Rademacher Group Limited Phosphoglycan messengers and their medical uses
UA72612C2 (en) 2000-07-06 2005-03-15 Pyrido[2.3-d]pyrimidine and pyrimido[4.5-d]pyrimidine nucleoside analogues, prodrugs and method for inhibiting growth of neoplastic cells
US6448281B1 (en) 2000-07-06 2002-09-10 Boehringer Ingelheim (Canada) Ltd. Viral polymerase inhibitors
CN1443190A (en) 2000-07-18 2003-09-17 里奥制药有限公司 Matrix metalloproteinase inhibitors
GB0017676D0 (en) 2000-07-19 2000-09-06 Angeletti P Ist Richerche Bio Inhibitors of viral polymerase
EE05366B1 (en) * 2000-07-21 2010-12-15 Gilead Sciences, Inc Prodrugs and Screening Methods for Phosphonate Nucleotide Analogs
AU2001292557A1 (en) 2000-08-22 2002-03-04 Ribapharm Inc. Methods of drug delivery to hepatocytes and treatment of flaviviridae infections
US20030008841A1 (en) 2000-08-30 2003-01-09 Rene Devos Anti-HCV nucleoside derivatives
AU2001282528A1 (en) 2000-09-01 2002-03-22 Shionogi And Co., Ltd. Compounds having anti-hepatitis c virus effect
CN1646141B (en) 2000-10-18 2014-06-25 吉利德制药有限责任公司 Modified nucleosides for treatment of viral infections and abnormal cellular proliferation
US20040014696A1 (en) 2000-12-07 2004-01-22 Johnson Lau Specificity in treatment of diseases
HUP0303436A2 (en) 2000-12-12 2004-01-28 Schering Corp. Diaryl peptides as ns3-serine protease inhibitors of hepatitis c virus, pharmaceutical compositions containing them and their use
US6653295B2 (en) 2000-12-13 2003-11-25 Bristol-Myers Squibb Company Inhibitors of hepatitis C virus NS3 protease
BR0116221A (en) 2000-12-15 2005-09-13 Pharmasset Ltd Antiviral agents for treatment of flaviviridae infections
CA2437209A1 (en) 2000-12-26 2002-07-04 Mitsubishi Pharma Corporation Remedies for hepatitis c
KR100828453B1 (en) 2001-01-22 2008-05-13 머크 앤드 캄파니 인코포레이티드 Nucleoside derivatives as inhibitors of RNA-dependent RNA viral polymerase
EP1390029B1 (en) 2001-04-11 2009-12-16 Idenix (Cayman) Limited Phenylindoles for the treatment of hiv
GB0114286D0 (en) 2001-06-12 2001-08-01 Hoffmann La Roche Nucleoside Derivatives
GB2378524A (en) 2001-08-08 2003-02-12 Bookham Technology Plc Intergrated optic device
GB0119369D0 (en) 2001-08-08 2001-10-03 Bookham Technology Plc Optic system
US20040077563A1 (en) 2001-08-21 2004-04-22 Johnson Lau Methods of drug delivery to hepatocytes and treatment of flaviviridae infections
US20030232760A1 (en) 2001-09-21 2003-12-18 Merck & Co., Inc. Conjugates useful in the treatment of prostate cancer
JP2005512516A (en) 2001-09-24 2005-05-12 メルク エンド カムパニー インコーポレーテッド Methods for screening and selecting combinations of statin drugs
EP1435974A4 (en) 2001-09-28 2006-09-06 Idenix Cayman Ltd Methods and compositions for treating hepatitis c virus using 4'-modified nucleosides
WO2003026675A1 (en) 2001-09-28 2003-04-03 Idenix (Cayman) Limited Methods and compositions for treating flaviviruses and pestiviruses using 4'-modified nucleoside
US20030072451A1 (en) 2001-10-16 2003-04-17 Pimentel Roberto J. Method and apparatus for securely transferring wireless data
JPWO2003034709A1 (en) 2001-10-17 2005-02-10 ナルテック株式会社 Decompression method and data processing apparatus
WO2003037908A1 (en) 2001-10-31 2003-05-08 Ribapharm Inc. Antiviral combination therapy and compositions
AU2002357876A1 (en) 2001-12-17 2003-06-30 Ribapharm Inc. Nucleoside analog libraries and compounds
AU2002340387A1 (en) 2001-12-17 2003-06-30 Ribapharm Inc. Cytidine libraries and compounds synthesized by solid-phase combinatorial strategies
AU2002353165A1 (en) 2001-12-17 2003-06-30 Ribapharm Inc. Deazapurine nucleoside libraries and compounds
AU2002359732A1 (en) 2001-12-17 2003-06-30 Ribapharm Inc. Substituted purine nucleoside libraries and compounds by solid-phase combinatorial strategies
WO2003051898A1 (en) 2001-12-17 2003-06-26 Ribapharm Inc. Unusual nucleoside libraries, compounds, and preferred uses as antiviral and anticancer agents
AU2002349905A1 (en) 2001-12-17 2003-06-30 Ribapharm Inc. Nucleoside libraries and compounds by mcc combinatorial strategies on solid support
US7261704B2 (en) 2001-12-28 2007-08-28 Terumo Kabushiki Kaisha Syringe
EP1572705A2 (en) 2002-01-17 2005-09-14 Ribapharm, Inc. Sugar modified nucleosides as viral replication inhibitors
WO2003068244A1 (en) 2002-02-13 2003-08-21 Merck & Co., Inc. Methods of inhibiting orthopoxvirus replication with nucleoside compounds
US20040063658A1 (en) 2002-05-06 2004-04-01 Roberts Christopher Don Nucleoside derivatives for treating hepatitis C virus infection
JP4476811B2 (en) 2002-05-13 2010-06-09 メタバシス・セラピューティクス・インコーポレイテッド New phosphonic acid prodrugs of PMEA and its analogs
CA2485597C (en) 2002-05-13 2013-07-02 Metabasis Therapeutics, Inc. Process for preparation of cyclic prodrugs of pmea and pmpa
EP1572945A2 (en) 2002-06-27 2005-09-14 Merck & Co., Inc. Nucleoside derivatives as inhibitors of rna-dependent rna viral polymerase
BR0312278A (en) 2002-06-28 2007-06-19 Idenix Cayman Ltd 2'-c-methyl-3'-o-1-valine ribofuranosyl cytidine ester for treatment of flaviviridae infections
JP5087211B2 (en) 2002-06-28 2012-12-05 イデニクス(ケイマン)リミテツド 2 'and 3'-nucleoside prodrugs for the treatment of flavivirus infection
US7608600B2 (en) 2002-06-28 2009-10-27 Idenix Pharmaceuticals, Inc. Modified 2′ and 3′-nucleoside prodrugs for treating Flaviviridae infections
AU2003247084B9 (en) 2002-06-28 2018-07-26 Centre National De La Recherche Scientifique Modified 2' and 3'-nucleoside prodrugs for treating flaviridae infections
US20040067877A1 (en) 2002-08-01 2004-04-08 Schinazi Raymond F. 2', 3'-Dideoxynucleoside analogues for the treatment or prevention of Flaviviridae infections
NZ538457A (en) 2002-09-30 2008-04-30 Genelabs Tech Inc Nucleoside derivatives for treating hepatitis C virus infection
US7094768B2 (en) 2002-09-30 2006-08-22 Genelabs Technologies, Inc. Nucleoside derivatives for treating hepatitis C virus infection
DE60329211D1 (en) 2002-10-31 2009-10-22 Metabasis Therapeutics Inc CYTARABIN MONOPHOSPHATE PRODRUGS
RU2005118421A (en) 2002-11-15 2006-01-20 Айденикс (Кайман) Лимитед (Ky) 2'-BRANCHED NUCLEOSIDES AND MUTATION FLAVIVIRIDAE
TWI332507B (en) 2002-11-19 2010-11-01 Hoffmann La Roche Antiviral nucleoside derivatives
TWI294882B (en) 2002-12-09 2008-03-21 Hoffmann La Roche Anhydrous crystalline azido cytosine hemisulfate derivative
PT3521297T (en) 2003-05-30 2022-03-18 Gilead Pharmasset Llc Modified fluorinated nucleoside analogues
ES2351603T3 (en) 2003-07-25 2011-02-08 Idenix Pharmaceuticals, Inc. PURINE NUCLEOSID ANALOGS FOR THE TREATMENT OF DISEASES CAUSED BY FLAVIRIDAE INCLUDING HEPATITIS C.
TW200510425A (en) 2003-08-13 2005-03-16 Japan Tobacco Inc Nitrogen-containing fused ring compound and use thereof as HIV integrase inhibitor
US7491794B2 (en) 2003-10-14 2009-02-17 Intermune, Inc. Macrocyclic compounds as inhibitors of viral replication
US7151089B2 (en) 2003-10-27 2006-12-19 Genelabs Technologies, Inc. Nucleoside compounds for treating viral infections
CA2542776A1 (en) 2003-10-27 2005-05-12 Genelabs Technologies, Inc. Nucleoside compounds for treating viral infections
ES2405404T3 (en) 2003-10-29 2013-05-31 Wyeth Llc Pharmaceutical sustained release compositions comprising aplindore and its derivatives
EP1704856A4 (en) 2003-12-26 2009-08-19 Kyowa Hakko Kirin Co Ltd Hsp90 family protein inhibitor
ES2389602T3 (en) 2003-12-30 2012-10-29 Gilead Sciences, Inc. Nucleoside phosphonates and analogues thereof for the treatment of HPV infections
JP5142716B2 (en) * 2004-06-08 2013-02-13 リガンド・ファーマシューティカルズ・インコーポレイテッド Lewis acid mediated synthesis of cyclic esters
JP2008508319A (en) 2004-07-29 2008-03-21 メタバシス・セラピューティクス・インコーポレイテッド Novel nucleoside derivatives
US7206468B2 (en) 2004-08-17 2007-04-17 Huang Hung-Chia Broad-band fiber-optic wave plates
JP5416403B2 (en) 2005-06-24 2014-02-12 レツク・フアーマシユーテイカルズ・デー・デー Process for the preparation of pure amorphous rosuvastatin calcium
US8003625B2 (en) * 2005-06-29 2011-08-23 Threshold Pharmaceuticals, Inc. Phosphoramidate alkylator prodrugs
US20070078111A1 (en) 2005-07-19 2007-04-05 Gabor Tigyi LPA2 receptor agonist inhibitors of CFTR
WO2007017117A1 (en) 2005-07-28 2007-02-15 Lek Pharmaceuticals D.D. Process for the synthesis of rosuvastatin calcium
WO2007022073A2 (en) * 2005-08-12 2007-02-22 Merck & Co., Inc. Novel 2'-c-methyl and 4'-c-methyl nucleoside derivatives
EP1928475B1 (en) 2005-08-15 2018-05-23 Riboscience LLC Antiviral phosphoramidates of 4'-c-azido-substituted pronucleotides
JP4954573B2 (en) 2006-02-28 2012-06-20 オリンパス株式会社 Endoscope system
HUE027872T2 (en) 2006-05-03 2016-11-28 Msn Laboratories Private Ltd Novel process for statins and its pharmaceutically acceptable salts thereof
US7795291B2 (en) * 2006-07-07 2010-09-14 Bristol-Myers Squibb Company Substituted acid derivatives useful as anti-atherosclerotic, anti-dyslipidemic, anti-diabetic and anti-obesity agents and method
WO2009023631A1 (en) 2007-08-10 2009-02-19 Basil Rigas Anti-inflammatory compounds and uses thereof
IT1391812B1 (en) 2008-07-29 2012-01-27 Maria Grazia Rimoli KETOROLAC GALACTOSILATE PROFARM FOR ANALGESIC AND ANTI-INFLAMMATORY USE WITH BETTER PHARMACOKINETIC CHARACTERISTICS AND TOXICOLOGICAL PROFILE OF THE STARTING DRUG
WO2010042600A2 (en) 2008-10-08 2010-04-15 The Uab Research Foundation Photo-activatable therapeutic agents and methods of using
JPWO2010047296A1 (en) 2008-10-20 2012-03-22 株式会社カネカ Method for producing novel pyrimidine derivative and intermediate of HMG-CoA reductase inhibitor
WO2010065760A1 (en) * 2008-12-04 2010-06-10 Exelixis, Inc. Imidazo [1,2a] pyridine derivatives, their use as s1p1 agonists and methods for their production
KR101091130B1 (en) 2009-01-29 2011-12-09 (주)유케이케미팜 Precursive compound of rosuvastatin and manufacturing method for the precursive compound of rosuvastatin
GB0904100D0 (en) 2009-03-10 2009-04-22 Bradford Pharma Ltd Use of rosuvastatin lactols as medicaments
WO2010105048A1 (en) 2009-03-12 2010-09-16 Metabolic Solutions Development Company Thiazolidinedione analogues
SG10201406846XA (en) * 2009-10-26 2014-12-30 Warner Chilcott Co Llc Bisphosphonate compounds for treating bone metabolism disorders
CN102079726B (en) 2009-11-27 2013-04-03 上海医药工业研究院 Miazine compounds, intermediates of miazine compounds, preparation method of intermediates and miazine compounds as well as application of miazine compound
WO2011160974A2 (en) 2010-06-21 2011-12-29 Nicox S.A. Statin derivatives
WO2012038785A1 (en) 2010-09-21 2012-03-29 Biocon Limited Polymorphs of rosuvastatin acetonide calcium ((3r,5s,6e)-7-[4-(4- fluorophenyl)-6-isopropyl-2-(methanesulfonyl-methyl-amino)-pyrimn)in-5- yl)vinyl)-2,2-dimethyl-l,3-dioxan-4-yl) acetic acid calcium salt
US9095599B2 (en) * 2011-01-03 2015-08-04 Nanjing Molecular Research, Inc. O-(substituted benzyl) phosphoramidate compounds and therapeutic use
KR20140033446A (en) * 2011-05-19 2014-03-18 알에프에스 파마 엘엘씨 Purine monophosphate prodrugs for treatment of viral infections
KR101444693B1 (en) 2011-06-06 2014-09-26 아버 테라퓨틱스 엘엘씨 Acid―labile lipophilic prodrugs of cancer chemotherapeutic agents
WO2013171639A1 (en) 2012-05-15 2013-11-21 Novartis Ag Benzamide derivatives for inhibiting the activity of abl1, abl2 and bcr-abl1
US20140142052A1 (en) 2012-05-17 2014-05-22 Normoxys, Inc. Phosphorylated polyols, pyrophosphates, and derivatives thereof having biological activity
WO2013187978A1 (en) * 2012-06-16 2013-12-19 Nanjing Molecular Research, Inc. Double-liver-targeting phosphoramidate and phosphonoamidate prodrugs
US9326991B2 (en) 2012-09-14 2016-05-03 Ligand Pharmaceuticals, Inc. Nucleotide prodrug compounds and use
WO2014052896A1 (en) * 2012-09-28 2014-04-03 Tufts University Uridine diphosphate derivatives, prodrugs, compositions and uses thereof
US9850213B2 (en) 2013-11-25 2017-12-26 Jiangxi Boya Seehot Pharmaceutical Co., Ltd. Method for preparing rosuvastatin sodium
US10449210B2 (en) 2014-02-13 2019-10-22 Ligand Pharmaceuticals Inc. Prodrug compounds and their uses
EP3164136A4 (en) 2014-07-02 2018-04-04 Ligand Pharmaceuticals, Inc. Prodrug compounds and uses therof
WO2016130417A1 (en) 2015-02-11 2016-08-18 Omthera Pharmaceuticals Inc Omega-3 fatty acid prodrug compounds and uses thereof
CA3087932A1 (en) 2018-01-09 2019-07-18 Ligand Pharmaceuticals, Inc. Acetal compounds and therapeutic uses thereof

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6312662B1 (en) 1998-03-06 2001-11-06 Metabasis Therapeutics, Inc. Prodrugs phosphorus-containing compounds
US7205404B1 (en) 1999-03-05 2007-04-17 Metabasis Therapeutics, Inc. Phosphorus-containing prodrugs
WO2001019829A2 (en) * 1999-09-17 2001-03-22 Basf Aktiengesellschaft Pyrazolopyrimidines as therapeutic agents
US20060014740A1 (en) * 2003-11-18 2006-01-19 Miller Duane D Analogs exhibiting inhibition of cell proliferation, methods of making, and uses thereof
US7666855B2 (en) 2004-02-13 2010-02-23 Metabasis Therapeutics, Inc. 2′-C-methyl nucleoside derivatives
WO2008116156A2 (en) * 2007-03-21 2008-09-25 The Regents Of The University Of Michigan Methods and compositions for treating diseases and conditions associated with mitochondrial function
WO2009011850A2 (en) * 2007-07-16 2009-01-22 Abbott Laboratories Novel therapeutic compounds
WO2009073506A2 (en) 2007-11-29 2009-06-11 Metabasis Therapeutics Inc. Nucleoside prodrugs and uses thereof
US8063025B2 (en) 2007-11-29 2011-11-22 Metabasis Therapeutics, Inc. Nucleoside prodrugs and uses thereof
WO2011084402A1 (en) * 2009-12-21 2011-07-14 Merck Sharp & Dohme Corp. Tyrosine kinase inhibitors
WO2013074386A2 (en) * 2011-11-15 2013-05-23 Merck Sharp & Dohme Corp. Hcv ns3 protease inhibitors

Non-Patent Citations (12)

* Cited by examiner, † Cited by third party
Title
BERRY, M.N.FRIEND, D.S., J. CELL BIOL., vol. 43, 1969, pages 506 - 520
CRISTINA CRUZ-HERNANDEZ ET AL: "Benefits of Structured and Free Monoacylglycerols to Deliver Eicosapentaenoic (EPA) in a Model of Lipid Malabsorption", NUTRIENTS, vol. 4, no. 12, 21 November 2012 (2012-11-21), pages 1781 - 1793, XP055155476, DOI: 10.3390/nu4111781 *
DEWAZIERS ET AL., J PHARM EXP THER, vol. 253, 1990, pages 387
GROEN, A.K. ET AL., EUR. J. BIOCHEM, vol. 122, 1982, pages 87 - 93
J. E. STARRETT, JR. ET AL., J MED CHEM, vol. 37, 1994, pages 1857 - 1864
J. K. DICKSON ET AL., J MED CHEM, vol. 39, 1996, pages 661 - 664
J. RAUTIO ET AL., NATURE REVIEWS DRUG DISCOVERY, vol. 7, 2008, pages 255 - 270
KIM H O ET AL: "L-SS-(2S,4S)- AND L-A-(2S,4R)-DIOXOLANYL NUCLEOSIDES AS POTENTIAL ANTI-HIV AGENTS: ASYMMETRIC SYNTHESIS AND STRUCTURE-ACTIVITY RELATIONSHIP", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 36, no. 5, 1 January 1993 (1993-01-01), pages 519 - 528, XP000887154, ISSN: 0022-2623, DOI: 10.1021/JM00057A001 *
M. D. ERION ET AL., J PHARM EXP THER, vol. 312, 2005, pages 554 - 60
X. J. ZHOU ET AL., EASL MEETING POSTER #966, 2009
XIAO MEI ET AL: "Hollow mesoporous silica nanoparticles conjugated with pH-sensitive amphiphilic diblock polymer for controlled drug release", MICROPOROUS AND MESOPOROUS MATERIALS, ELSEVIER, AMSTERDAM, NL, vol. 152, 6 December 2011 (2011-12-06), pages 16 - 24, XP028443325, ISSN: 1387-1811, [retrieved on 20111213], DOI: 10.1016/J.MICROMESO.2011.12.015 *
YURI WEDMID ET AL: "Long-chain stereomeric 2-alkyl-4-methoxycarbonyl-1,3-dioxolanes in glycerol acetal synthesis", JOURNAL OF ORGANIC CHEMISTRY, vol. 42, no. 22, 1 October 1977 (1977-10-01), US, pages 3624 - 3626, XP055659449, ISSN: 0022-3263, DOI: 10.1021/jo00442a040 *

Also Published As

Publication number Publication date
CN114404427A (en) 2022-04-29
WO2015123352A1 (en) 2015-08-20
AU2015217221A1 (en) 2016-08-11
CN106061984A (en) 2016-10-26
WO2015123352A9 (en) 2016-09-09
JP2022009127A (en) 2022-01-14
CA2937548C (en) 2022-10-25
AU2015217221A9 (en) 2016-12-15
EP3105238A4 (en) 2017-11-08
CA2937548A1 (en) 2015-08-20
US10449210B2 (en) 2019-10-22
EP3105238A1 (en) 2016-12-21
JP2017512183A (en) 2017-05-18
JP2020073466A (en) 2020-05-14
AU2021225211A1 (en) 2021-09-30
US20190388452A1 (en) 2019-12-26
AU2020200786A1 (en) 2020-02-20
US11278559B2 (en) 2022-03-22
US20170056429A1 (en) 2017-03-02

Similar Documents

Publication Publication Date Title
US11278559B2 (en) Prodrug compounds and their uses
US10150788B2 (en) Prodrug compounds and uses thereof
AU2019207626B2 (en) Phosphor(n)amidatacetal and phosph(on)atalcetal compounds
TWI838355B (en) 5-fluorouracil compounds
WO2006091905A1 (en) Bicyclo (3.1.0) hexane derivatives as antiviral compounds
EA019559B1 (en) Antiviral compounds, in particular that inhibitis hiv, pharmaceutical composition based thereon and method for inhibiting using same
US20180044368A1 (en) Gemcitabine derivatives
CN115768778A (en) Benzyloxyphosphoric acid (phosphonate) compound
CN106661060A (en) Phenanthroline phosphonic acid derivative and preparation method therefor and application thereof
WO2022245584A1 (en) Unnatural configuration nucleotide prodrug compounds
RU2796403C2 (en) Phosphorus(n)amidateacetal and phosph(oh)atacetal compounds

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN PUBLISHED

AC Divisional application: reference to earlier application

Ref document number: 3105238

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20200914

RBV Designated contracting states (corrected)

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40026405

Country of ref document: HK

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20220120

RAP3 Party data changed (applicant data changed or rights of an application transferred)

Owner name: LIGAND PHARMACEUTICALS, INC.